WO2007005635A2 - Mitotic spindle protein aspm as a diagnostic marker for neoplasia and uses therefor - Google Patents
Mitotic spindle protein aspm as a diagnostic marker for neoplasia and uses therefor Download PDFInfo
- Publication number
- WO2007005635A2 WO2007005635A2 PCT/US2006/025640 US2006025640W WO2007005635A2 WO 2007005635 A2 WO2007005635 A2 WO 2007005635A2 US 2006025640 W US2006025640 W US 2006025640W WO 2007005635 A2 WO2007005635 A2 WO 2007005635A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- aspm
- cell
- nucleic acid
- expression
- neoplasia
- Prior art date
Links
- 206010028980 Neoplasm Diseases 0.000 title claims description 151
- 108090000623 proteins and genes Proteins 0.000 title claims description 131
- 230000009826 neoplastic cell growth Effects 0.000 title claims description 111
- 102000004169 proteins and genes Human genes 0.000 title claims description 109
- 230000000394 mitotic effect Effects 0.000 title description 18
- 239000003550 marker Substances 0.000 title description 2
- 230000014509 gene expression Effects 0.000 claims abstract description 167
- 102000039446 nucleic acids Human genes 0.000 claims abstract description 165
- 108020004707 nucleic acids Proteins 0.000 claims abstract description 164
- 150000007523 nucleic acids Chemical class 0.000 claims abstract description 161
- 101000900939 Homo sapiens Abnormal spindle-like microcephaly-associated protein Proteins 0.000 claims abstract description 88
- 230000001965 increasing effect Effects 0.000 claims abstract description 22
- 230000001613 neoplastic effect Effects 0.000 claims abstract description 9
- 102100022117 Abnormal spindle-like microcephaly-associated protein Human genes 0.000 claims abstract 56
- 238000000034 method Methods 0.000 claims description 174
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 173
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 164
- 229920001184 polypeptide Polymers 0.000 claims description 157
- 210000004027 cell Anatomy 0.000 claims description 148
- 150000001875 compounds Chemical class 0.000 claims description 109
- 239000012634 fragment Substances 0.000 claims description 65
- 239000000523 sample Substances 0.000 claims description 59
- 210000001519 tissue Anatomy 0.000 claims description 54
- 230000004075 alteration Effects 0.000 claims description 37
- 230000027455 binding Effects 0.000 claims description 37
- -1 ASPM nucleic acid Chemical class 0.000 claims description 28
- 239000013598 vector Substances 0.000 claims description 28
- 108091032973 (ribonucleotides)n+m Proteins 0.000 claims description 27
- 230000004071 biological effect Effects 0.000 claims description 27
- 238000001514 detection method Methods 0.000 claims description 22
- 238000002493 microarray Methods 0.000 claims description 21
- 108010041952 Calmodulin Proteins 0.000 claims description 19
- 102000000584 Calmodulin Human genes 0.000 claims description 19
- 238000009396 hybridization Methods 0.000 claims description 19
- 230000000694 effects Effects 0.000 claims description 17
- 102000040650 (ribonucleotides)n+m Human genes 0.000 claims description 15
- 208000037051 Chromosomal Instability Diseases 0.000 claims description 15
- 239000004055 small Interfering RNA Substances 0.000 claims description 14
- 108020004459 Small interfering RNA Proteins 0.000 claims description 13
- 210000000481 breast Anatomy 0.000 claims description 13
- 230000004663 cell proliferation Effects 0.000 claims description 13
- 239000003153 chemical reaction reagent Substances 0.000 claims description 13
- 238000000338 in vitro Methods 0.000 claims description 13
- 238000001727 in vivo Methods 0.000 claims description 13
- 230000000692 anti-sense effect Effects 0.000 claims description 12
- 239000008194 pharmaceutical composition Substances 0.000 claims description 12
- 210000005170 neoplastic cell Anatomy 0.000 claims description 11
- 239000002773 nucleotide Substances 0.000 claims description 11
- 125000003729 nucleotide group Chemical group 0.000 claims description 11
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 10
- 239000012472 biological sample Substances 0.000 claims description 10
- 230000000295 complement effect Effects 0.000 claims description 10
- 230000007423 decrease Effects 0.000 claims description 10
- 239000013604 expression vector Substances 0.000 claims description 10
- 230000002401 inhibitory effect Effects 0.000 claims description 10
- 210000004962 mammalian cell Anatomy 0.000 claims description 10
- 239000000758 substrate Substances 0.000 claims description 10
- 210000001550 testis Anatomy 0.000 claims description 10
- 239000000090 biomarker Substances 0.000 claims description 9
- 238000003745 diagnosis Methods 0.000 claims description 9
- 239000007787 solid Substances 0.000 claims description 9
- 108020001507 fusion proteins Proteins 0.000 claims description 8
- 102000037865 fusion proteins Human genes 0.000 claims description 8
- 210000005260 human cell Anatomy 0.000 claims description 8
- 210000004072 lung Anatomy 0.000 claims description 8
- 210000001672 ovary Anatomy 0.000 claims description 8
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 8
- 238000012544 monitoring process Methods 0.000 claims description 7
- 238000013518 transcription Methods 0.000 claims description 7
- 230000035897 transcription Effects 0.000 claims description 7
- 238000006243 chemical reaction Methods 0.000 claims description 5
- 238000009007 Diagnostic Kit Methods 0.000 claims description 4
- 108700008625 Reporter Genes Proteins 0.000 claims description 4
- 239000003463 adsorbent Substances 0.000 claims description 4
- 238000010324 immunological assay Methods 0.000 claims description 4
- 210000002307 prostate Anatomy 0.000 claims description 4
- 238000013519 translation Methods 0.000 claims description 4
- 238000011256 aggressive treatment Methods 0.000 claims description 3
- 238000010837 poor prognosis Methods 0.000 claims description 3
- 238000004393 prognosis Methods 0.000 claims description 3
- 238000000018 DNA microarray Methods 0.000 claims description 2
- 238000007824 enzymatic assay Methods 0.000 claims description 2
- 238000003127 radioimmunoassay Methods 0.000 claims description 2
- 238000011269 treatment regimen Methods 0.000 claims description 2
- ZPCCSZFPOXBNDL-ZSTSFXQOSA-N [(4r,5s,6s,7r,9r,10r,11e,13e,16r)-6-[(2s,3r,4r,5s,6r)-5-[(2s,4r,5s,6s)-4,5-dihydroxy-4,6-dimethyloxan-2-yl]oxy-4-(dimethylamino)-3-hydroxy-6-methyloxan-2-yl]oxy-10-[(2r,5s,6r)-5-(dimethylamino)-6-methyloxan-2-yl]oxy-5-methoxy-9,16-dimethyl-2-oxo-7-(2-oxoe Chemical compound O([C@H]1/C=C/C=C/C[C@@H](C)OC(=O)C[C@H]([C@@H]([C@H]([C@@H](CC=O)C[C@H]1C)O[C@H]1[C@@H]([C@H]([C@H](O[C@@H]2O[C@@H](C)[C@H](O)[C@](C)(O)C2)[C@@H](C)O1)N(C)C)O)OC)OC(C)=O)[C@H]1CC[C@H](N(C)C)[C@@H](C)O1 ZPCCSZFPOXBNDL-ZSTSFXQOSA-N 0.000 claims 23
- 239000000203 mixture Substances 0.000 abstract description 21
- 238000002560 therapeutic procedure Methods 0.000 abstract description 9
- 238000002405 diagnostic procedure Methods 0.000 abstract description 6
- 235000018102 proteins Nutrition 0.000 description 104
- 235000001014 amino acid Nutrition 0.000 description 48
- 241000282414 Homo sapiens Species 0.000 description 46
- 229940024606 amino acid Drugs 0.000 description 46
- 150000001413 amino acids Chemical class 0.000 description 45
- 102000040430 polynucleotide Human genes 0.000 description 34
- 108091033319 polynucleotide Proteins 0.000 description 34
- 239000002157 polynucleotide Substances 0.000 description 34
- 102000056090 human ASPM Human genes 0.000 description 31
- 241000699666 Mus <mouse, genus> Species 0.000 description 29
- 238000003556 assay Methods 0.000 description 27
- 201000011510 cancer Diseases 0.000 description 22
- 238000011282 treatment Methods 0.000 description 21
- 238000010240 RT-PCR analysis Methods 0.000 description 20
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 20
- 238000004458 analytical method Methods 0.000 description 20
- 108020004414 DNA Proteins 0.000 description 19
- 230000006870 function Effects 0.000 description 19
- 239000000284 extract Substances 0.000 description 16
- 238000003752 polymerase chain reaction Methods 0.000 description 16
- 108091028043 Nucleic acid sequence Proteins 0.000 description 15
- 108091034117 Oligonucleotide Proteins 0.000 description 15
- 108010029485 Protein Isoforms Proteins 0.000 description 15
- 102000001708 Protein Isoforms Human genes 0.000 description 15
- 210000001072 colon Anatomy 0.000 description 14
- 239000000126 substance Substances 0.000 description 14
- 238000013459 approach Methods 0.000 description 13
- 210000004556 brain Anatomy 0.000 description 13
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 13
- 238000010561 standard procedure Methods 0.000 description 12
- 206010061535 Ovarian neoplasm Diseases 0.000 description 11
- 201000010099 disease Diseases 0.000 description 11
- 230000002163 immunogen Effects 0.000 description 11
- 208000004141 microcephaly Diseases 0.000 description 11
- 239000001509 sodium citrate Substances 0.000 description 11
- 230000001225 therapeutic effect Effects 0.000 description 11
- HRXKRNGNAMMEHJ-UHFFFAOYSA-K trisodium citrate Chemical compound [Na+].[Na+].[Na+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O HRXKRNGNAMMEHJ-UHFFFAOYSA-K 0.000 description 11
- 229940038773 trisodium citrate Drugs 0.000 description 11
- 102000007469 Actins Human genes 0.000 description 10
- 108010085238 Actins Proteins 0.000 description 10
- 108700024394 Exon Proteins 0.000 description 10
- ZHNUHDYFZUAESO-UHFFFAOYSA-N Formamide Chemical compound NC=O ZHNUHDYFZUAESO-UHFFFAOYSA-N 0.000 description 10
- 101100217298 Mus musculus Aspm gene Proteins 0.000 description 10
- 125000000539 amino acid group Chemical group 0.000 description 10
- 210000003169 central nervous system Anatomy 0.000 description 10
- 239000002299 complementary DNA Substances 0.000 description 10
- 230000000875 corresponding effect Effects 0.000 description 10
- 239000000463 material Substances 0.000 description 10
- 239000012528 membrane Substances 0.000 description 10
- 239000011780 sodium chloride Substances 0.000 description 10
- 206010033128 Ovarian cancer Diseases 0.000 description 9
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 9
- 230000003993 interaction Effects 0.000 description 9
- 208000032839 leukemia Diseases 0.000 description 9
- 210000004185 liver Anatomy 0.000 description 9
- 238000004519 manufacturing process Methods 0.000 description 9
- 201000001441 melanoma Diseases 0.000 description 9
- 108020004999 messenger RNA Proteins 0.000 description 9
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 9
- 230000001105 regulatory effect Effects 0.000 description 9
- 101150034094 ASPM gene Proteins 0.000 description 8
- 241000124008 Mammalia Species 0.000 description 8
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 8
- 208000035269 cancer or benign tumor Diseases 0.000 description 8
- 238000003786 synthesis reaction Methods 0.000 description 8
- 238000005406 washing Methods 0.000 description 8
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 7
- 241000288906 Primates Species 0.000 description 7
- 208000002495 Uterine Neoplasms Diseases 0.000 description 7
- 210000003793 centrosome Anatomy 0.000 description 7
- 230000008859 change Effects 0.000 description 7
- 239000003814 drug Substances 0.000 description 7
- 230000002611 ovarian Effects 0.000 description 7
- 210000002784 stomach Anatomy 0.000 description 7
- 108700037179 Drosophila asp Proteins 0.000 description 6
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 6
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 6
- 241001529936 Murinae Species 0.000 description 6
- 108700026244 Open Reading Frames Proteins 0.000 description 6
- 230000000118 anti-neoplastic effect Effects 0.000 description 6
- 230000006907 apoptotic process Effects 0.000 description 6
- 230000001580 bacterial effect Effects 0.000 description 6
- 239000011324 bead Substances 0.000 description 6
- 210000004748 cultured cell Anatomy 0.000 description 6
- 238000012217 deletion Methods 0.000 description 6
- 230000037430 deletion Effects 0.000 description 6
- 230000011278 mitosis Effects 0.000 description 6
- 230000035772 mutation Effects 0.000 description 6
- 238000011160 research Methods 0.000 description 6
- 238000006467 substitution reaction Methods 0.000 description 6
- 206010046766 uterine cancer Diseases 0.000 description 6
- 238000001262 western blot Methods 0.000 description 6
- 241000255581 Drosophila <fruit fly, genus> Species 0.000 description 5
- 102000004190 Enzymes Human genes 0.000 description 5
- 108090000790 Enzymes Proteins 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- 102000003505 Myosin Human genes 0.000 description 5
- 108060008487 Myosin Proteins 0.000 description 5
- 230000002159 abnormal effect Effects 0.000 description 5
- 230000015572 biosynthetic process Effects 0.000 description 5
- 230000032823 cell division Effects 0.000 description 5
- 238000012512 characterization method Methods 0.000 description 5
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 5
- 229940088598 enzyme Drugs 0.000 description 5
- 230000001605 fetal effect Effects 0.000 description 5
- 210000002216 heart Anatomy 0.000 description 5
- 210000004408 hybridoma Anatomy 0.000 description 5
- 238000012423 maintenance Methods 0.000 description 5
- 210000001161 mammalian embryo Anatomy 0.000 description 5
- 230000004048 modification Effects 0.000 description 5
- 238000012986 modification Methods 0.000 description 5
- 210000002569 neuron Anatomy 0.000 description 5
- 239000002953 phosphate buffered saline Substances 0.000 description 5
- 239000013612 plasmid Substances 0.000 description 5
- 230000002062 proliferating effect Effects 0.000 description 5
- 230000035755 proliferation Effects 0.000 description 5
- 238000000746 purification Methods 0.000 description 5
- 238000003753 real-time PCR Methods 0.000 description 5
- 230000002829 reductive effect Effects 0.000 description 5
- 150000003839 salts Chemical class 0.000 description 5
- 238000012216 screening Methods 0.000 description 5
- 238000007423 screening assay Methods 0.000 description 5
- 238000012163 sequencing technique Methods 0.000 description 5
- 150000003384 small molecules Chemical class 0.000 description 5
- 210000001685 thyroid gland Anatomy 0.000 description 5
- 210000003932 urinary bladder Anatomy 0.000 description 5
- 206010003445 Ascites Diseases 0.000 description 4
- BHPQYMZQTOCNFJ-UHFFFAOYSA-N Calcium cation Chemical compound [Ca+2] BHPQYMZQTOCNFJ-UHFFFAOYSA-N 0.000 description 4
- 238000002965 ELISA Methods 0.000 description 4
- 102000029749 Microtubule Human genes 0.000 description 4
- 108091022875 Microtubule Proteins 0.000 description 4
- 241000700159 Rattus Species 0.000 description 4
- 108020004511 Recombinant DNA Proteins 0.000 description 4
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 4
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 4
- 241000700605 Viruses Species 0.000 description 4
- 238000001042 affinity chromatography Methods 0.000 description 4
- 125000000217 alkyl group Chemical group 0.000 description 4
- 230000033228 biological regulation Effects 0.000 description 4
- 230000002759 chromosomal effect Effects 0.000 description 4
- 238000010367 cloning Methods 0.000 description 4
- 238000011161 development Methods 0.000 description 4
- 239000003623 enhancer Substances 0.000 description 4
- 210000003754 fetus Anatomy 0.000 description 4
- 238000005194 fractionation Methods 0.000 description 4
- 230000004927 fusion Effects 0.000 description 4
- 239000000499 gel Substances 0.000 description 4
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 4
- 230000036541 health Effects 0.000 description 4
- 238000004128 high performance liquid chromatography Methods 0.000 description 4
- 238000003018 immunoassay Methods 0.000 description 4
- 230000001939 inductive effect Effects 0.000 description 4
- 238000003780 insertion Methods 0.000 description 4
- 230000037431 insertion Effects 0.000 description 4
- 210000003734 kidney Anatomy 0.000 description 4
- 210000004688 microtubule Anatomy 0.000 description 4
- 230000001177 retroviral effect Effects 0.000 description 4
- 241000894007 species Species 0.000 description 4
- 238000010186 staining Methods 0.000 description 4
- 238000012360 testing method Methods 0.000 description 4
- 210000004291 uterus Anatomy 0.000 description 4
- 239000003981 vehicle Substances 0.000 description 4
- 230000002861 ventricular Effects 0.000 description 4
- 230000003612 virological effect Effects 0.000 description 4
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 3
- 102000053602 DNA Human genes 0.000 description 3
- SHIBSTMRCDJXLN-UHFFFAOYSA-N Digoxigenin Natural products C1CC(C2C(C3(C)CCC(O)CC3CC2)CC2O)(O)C2(C)C1C1=CC(=O)OC1 SHIBSTMRCDJXLN-UHFFFAOYSA-N 0.000 description 3
- 241000588724 Escherichia coli Species 0.000 description 3
- 239000004471 Glycine Substances 0.000 description 3
- 108060003951 Immunoglobulin Proteins 0.000 description 3
- 241000829100 Macaca mulatta polyomavirus 1 Species 0.000 description 3
- 108010020004 Microtubule-Associated Proteins Proteins 0.000 description 3
- 102000009664 Microtubule-Associated Proteins Human genes 0.000 description 3
- 108010028299 Myosin Type V Proteins 0.000 description 3
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 3
- 235000014680 Saccharomyces cerevisiae Nutrition 0.000 description 3
- 108091027967 Small hairpin RNA Proteins 0.000 description 3
- 102100030409 Unconventional myosin-Va Human genes 0.000 description 3
- 230000001154 acute effect Effects 0.000 description 3
- 239000000427 antigen Substances 0.000 description 3
- 108091007433 antigens Proteins 0.000 description 3
- 102000036639 antigens Human genes 0.000 description 3
- 238000001574 biopsy Methods 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 229910052799 carbon Inorganic materials 0.000 description 3
- 230000003833 cell viability Effects 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 230000002490 cerebral effect Effects 0.000 description 3
- 210000003679 cervix uteri Anatomy 0.000 description 3
- 239000003795 chemical substances by application Substances 0.000 description 3
- 239000013599 cloning vector Substances 0.000 description 3
- 230000001054 cortical effect Effects 0.000 description 3
- 229960004397 cyclophosphamide Drugs 0.000 description 3
- 230000034994 death Effects 0.000 description 3
- 231100000517 death Toxicity 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- QONQRTHLHBTMGP-UHFFFAOYSA-N digitoxigenin Natural products CC12CCC(C3(CCC(O)CC3CC3)C)C3C11OC1CC2C1=CC(=O)OC1 QONQRTHLHBTMGP-UHFFFAOYSA-N 0.000 description 3
- 210000003238 esophagus Anatomy 0.000 description 3
- 239000000835 fiber Substances 0.000 description 3
- 238000001415 gene therapy Methods 0.000 description 3
- 239000011521 glass Substances 0.000 description 3
- 230000012010 growth Effects 0.000 description 3
- 238000003119 immunoblot Methods 0.000 description 3
- 102000018358 immunoglobulin Human genes 0.000 description 3
- 238000007901 in situ hybridization Methods 0.000 description 3
- 210000001165 lymph node Anatomy 0.000 description 3
- 239000003068 molecular probe Substances 0.000 description 3
- 230000001537 neural effect Effects 0.000 description 3
- 210000000496 pancreas Anatomy 0.000 description 3
- 230000026731 phosphorylation Effects 0.000 description 3
- 238000006366 phosphorylation reaction Methods 0.000 description 3
- 238000006116 polymerization reaction Methods 0.000 description 3
- 230000004481 post-translational protein modification Effects 0.000 description 3
- 239000002243 precursor Substances 0.000 description 3
- 230000001603 reducing effect Effects 0.000 description 3
- 239000000243 solution Substances 0.000 description 3
- 235000000346 sugar Nutrition 0.000 description 3
- 238000012546 transfer Methods 0.000 description 3
- 230000009466 transformation Effects 0.000 description 3
- AVWQQPYHYQKEIZ-UHFFFAOYSA-K trisodium;2-dodecylbenzenesulfonate;3-dodecylbenzenesulfonate;4-dodecylbenzenesulfonate Chemical compound [Na+].[Na+].[Na+].CCCCCCCCCCCCC1=CC=C(S([O-])(=O)=O)C=C1.CCCCCCCCCCCCC1=CC=CC(S([O-])(=O)=O)=C1.CCCCCCCCCCCCC1=CC=CC=C1S([O-])(=O)=O AVWQQPYHYQKEIZ-UHFFFAOYSA-K 0.000 description 3
- 210000002700 urine Anatomy 0.000 description 3
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 2
- FDKXTQMXEQVLRF-ZHACJKMWSA-N (E)-dacarbazine Chemical compound CN(C)\N=N\c1[nH]cnc1C(N)=O FDKXTQMXEQVLRF-ZHACJKMWSA-N 0.000 description 2
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 2
- 229930024421 Adenine Natural products 0.000 description 2
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 2
- 239000004475 Arginine Substances 0.000 description 2
- 208000000848 Autosomal recessive primary microcephaly Diseases 0.000 description 2
- 241000894006 Bacteria Species 0.000 description 2
- 108050001630 Calponin homology domains Proteins 0.000 description 2
- 102000011398 Calponin homology domains Human genes 0.000 description 2
- 201000009030 Carcinoma Diseases 0.000 description 2
- DLGOEMSEDOSKAD-UHFFFAOYSA-N Carmustine Chemical compound ClCCNC(=O)N(N=O)CCCl DLGOEMSEDOSKAD-UHFFFAOYSA-N 0.000 description 2
- 238000003734 CellTiter-Glo Luminescent Cell Viability Assay Methods 0.000 description 2
- 108091026890 Coding region Proteins 0.000 description 2
- 108020004705 Codon Proteins 0.000 description 2
- 108010035532 Collagen Proteins 0.000 description 2
- 102000008186 Collagen Human genes 0.000 description 2
- 108091035707 Consensus sequence Proteins 0.000 description 2
- 150000008574 D-amino acids Chemical class 0.000 description 2
- 230000006820 DNA synthesis Effects 0.000 description 2
- 206010014759 Endometrial neoplasm Diseases 0.000 description 2
- 108091060211 Expressed sequence tag Proteins 0.000 description 2
- 108010008177 Fd immunoglobulins Proteins 0.000 description 2
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 2
- 241000287828 Gallus gallus Species 0.000 description 2
- 108010024636 Glutathione Proteins 0.000 description 2
- 102000005720 Glutathione transferase Human genes 0.000 description 2
- 108010070675 Glutathione transferase Proteins 0.000 description 2
- 241000238631 Hexapoda Species 0.000 description 2
- 208000017604 Hodgkin disease Diseases 0.000 description 2
- 208000010747 Hodgkins lymphoma Diseases 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 2
- 206010062717 Increased upper airway secretion Diseases 0.000 description 2
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 2
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 2
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 2
- LRQKBLKVPFOOQJ-YFKPBYRVSA-N L-norleucine Chemical compound CCCC[C@H]([NH3+])C([O-])=O LRQKBLKVPFOOQJ-YFKPBYRVSA-N 0.000 description 2
- GQYIWUVLTXOXAJ-UHFFFAOYSA-N Lomustine Chemical compound ClCCN(N=O)C(=O)NC1CCCCC1 GQYIWUVLTXOXAJ-UHFFFAOYSA-N 0.000 description 2
- 239000004472 Lysine Substances 0.000 description 2
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 2
- 241000699670 Mus sp. Species 0.000 description 2
- 108020004711 Nucleic Acid Probes Proteins 0.000 description 2
- 108700020796 Oncogene Proteins 0.000 description 2
- 239000002033 PVDF binder Substances 0.000 description 2
- 229930012538 Paclitaxel Natural products 0.000 description 2
- 108010033276 Peptide Fragments Proteins 0.000 description 2
- 102000007079 Peptide Fragments Human genes 0.000 description 2
- 108010043958 Peptoids Proteins 0.000 description 2
- 108091000080 Phosphotransferase Proteins 0.000 description 2
- 239000004952 Polyamide Substances 0.000 description 2
- 238000011529 RT qPCR Methods 0.000 description 2
- 101710137500 T7 RNA polymerase Proteins 0.000 description 2
- NKANXQFJJICGDU-QPLCGJKRSA-N Tamoxifen Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 NKANXQFJJICGDU-QPLCGJKRSA-N 0.000 description 2
- 208000024313 Testicular Neoplasms Diseases 0.000 description 2
- 206010057644 Testis cancer Diseases 0.000 description 2
- 102000004243 Tubulin Human genes 0.000 description 2
- 108090000704 Tubulin Proteins 0.000 description 2
- 241000700618 Vaccinia virus Species 0.000 description 2
- RJURFGZVJUQBHK-UHFFFAOYSA-N actinomycin D Natural products CC1OC(=O)C(C(C)C)N(C)C(=O)CN(C)C(=O)C2CCCN2C(=O)C(C(C)C)NC(=O)C1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)NC4C(=O)NC(C(N5CCCC5C(=O)N(C)CC(=O)N(C)C(C(C)C)C(=O)OC4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-UHFFFAOYSA-N 0.000 description 2
- 229960000643 adenine Drugs 0.000 description 2
- 208000009956 adenocarcinoma Diseases 0.000 description 2
- 239000011543 agarose gel Substances 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 230000031016 anaphase Effects 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 239000005557 antagonist Substances 0.000 description 2
- 229940034982 antineoplastic agent Drugs 0.000 description 2
- 239000002246 antineoplastic agent Substances 0.000 description 2
- 230000001640 apoptogenic effect Effects 0.000 description 2
- 239000007864 aqueous solution Substances 0.000 description 2
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 2
- 210000001124 body fluid Anatomy 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 210000004899 c-terminal region Anatomy 0.000 description 2
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 2
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 2
- 238000002512 chemotherapy Methods 0.000 description 2
- 238000004587 chromatography analysis Methods 0.000 description 2
- 210000000349 chromosome Anatomy 0.000 description 2
- 239000011248 coating agent Substances 0.000 description 2
- 238000000576 coating method Methods 0.000 description 2
- 229920001436 collagen Polymers 0.000 description 2
- 230000000052 comparative effect Effects 0.000 description 2
- 239000003184 complementary RNA Substances 0.000 description 2
- 238000011443 conventional therapy Methods 0.000 description 2
- 230000002596 correlated effect Effects 0.000 description 2
- 239000000287 crude extract Substances 0.000 description 2
- 230000002950 deficient Effects 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 210000002451 diencephalon Anatomy 0.000 description 2
- SHIBSTMRCDJXLN-KCZCNTNESA-N digoxigenin Chemical compound C1([C@@H]2[C@@]3([C@@](CC2)(O)[C@H]2[C@@H]([C@@]4(C)CC[C@H](O)C[C@H]4CC2)C[C@H]3O)C)=CC(=O)OC1 SHIBSTMRCDJXLN-KCZCNTNESA-N 0.000 description 2
- 208000035475 disorder Diseases 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 238000009509 drug development Methods 0.000 description 2
- 238000007876 drug discovery Methods 0.000 description 2
- 239000000975 dye Substances 0.000 description 2
- 210000004696 endometrium Anatomy 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- ZMMJGEGLRURXTF-UHFFFAOYSA-N ethidium bromide Chemical compound [Br-].C12=CC(N)=CC=C2C2=CC=C(N)C=C2[N+](CC)=C1C1=CC=CC=C1 ZMMJGEGLRURXTF-UHFFFAOYSA-N 0.000 description 2
- 229960005542 ethidium bromide Drugs 0.000 description 2
- 210000003527 eukaryotic cell Anatomy 0.000 description 2
- 238000010195 expression analysis Methods 0.000 description 2
- 239000012530 fluid Substances 0.000 description 2
- 239000007850 fluorescent dye Substances 0.000 description 2
- 229960002949 fluorouracil Drugs 0.000 description 2
- 238000013467 fragmentation Methods 0.000 description 2
- 238000006062 fragmentation reaction Methods 0.000 description 2
- 230000037433 frameshift Effects 0.000 description 2
- 230000000574 ganglionic effect Effects 0.000 description 2
- 238000001641 gel filtration chromatography Methods 0.000 description 2
- 230000002068 genetic effect Effects 0.000 description 2
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 2
- 235000004554 glutamine Nutrition 0.000 description 2
- 229960003180 glutathione Drugs 0.000 description 2
- 239000001963 growth medium Substances 0.000 description 2
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 2
- 125000000623 heterocyclic group Chemical group 0.000 description 2
- 229910052739 hydrogen Inorganic materials 0.000 description 2
- 239000001257 hydrogen Substances 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- 238000003384 imaging method Methods 0.000 description 2
- 238000003125 immunofluorescent labeling Methods 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- 229960000310 isoleucine Drugs 0.000 description 2
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 2
- 231100000225 lethality Toxicity 0.000 description 2
- 239000002502 liposome Substances 0.000 description 2
- 230000004807 localization Effects 0.000 description 2
- 210000002751 lymph Anatomy 0.000 description 2
- 239000012931 lyophilized formulation Substances 0.000 description 2
- 230000036210 malignancy Effects 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 229960004961 mechlorethamine Drugs 0.000 description 2
- HAWPXGHAZFHHAD-UHFFFAOYSA-N mechlorethamine Chemical compound ClCCN(C)CCCl HAWPXGHAZFHHAD-UHFFFAOYSA-N 0.000 description 2
- 239000002609 medium Substances 0.000 description 2
- 230000031864 metaphase Effects 0.000 description 2
- 238000000386 microscopy Methods 0.000 description 2
- 238000010369 molecular cloning Methods 0.000 description 2
- 230000009456 molecular mechanism Effects 0.000 description 2
- 229930014626 natural product Natural products 0.000 description 2
- 230000004766 neurogenesis Effects 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 239000002853 nucleic acid probe Substances 0.000 description 2
- 210000000056 organ Anatomy 0.000 description 2
- 230000008520 organization Effects 0.000 description 2
- 229960001592 paclitaxel Drugs 0.000 description 2
- 239000013610 patient sample Substances 0.000 description 2
- 208000026435 phlegm Diseases 0.000 description 2
- 102000020233 phosphotransferase Human genes 0.000 description 2
- 229920002647 polyamide Polymers 0.000 description 2
- 229920000642 polymer Polymers 0.000 description 2
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 201000001729 primary autosomal recessive microcephaly Diseases 0.000 description 2
- XOJVVFBFDXDTEG-UHFFFAOYSA-N pristane Chemical compound CC(C)CCCC(C)CCCC(C)CCCC(C)C XOJVVFBFDXDTEG-UHFFFAOYSA-N 0.000 description 2
- 210000001236 prokaryotic cell Anatomy 0.000 description 2
- 230000004853 protein function Effects 0.000 description 2
- 230000020978 protein processing Effects 0.000 description 2
- 238000001959 radiotherapy Methods 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 230000010076 replication Effects 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 210000003296 saliva Anatomy 0.000 description 2
- 210000002027 skeletal muscle Anatomy 0.000 description 2
- 210000003491 skin Anatomy 0.000 description 2
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 2
- 239000007790 solid phase Substances 0.000 description 2
- 210000000952 spleen Anatomy 0.000 description 2
- 238000001356 surgical procedure Methods 0.000 description 2
- RCINICONZNJXQF-MZXODVADSA-N taxol Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 description 2
- 201000003120 testicular cancer Diseases 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 239000013603 viral vector Substances 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- DIGQNXIGRZPYDK-WKSCXVIASA-N (2R)-6-amino-2-[[2-[[(2S)-2-[[2-[[(2R)-2-[[(2S)-2-[[(2R,3S)-2-[[2-[[(2S)-2-[[2-[[(2S)-2-[[(2S)-2-[[(2R)-2-[[(2S,3S)-2-[[(2R)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[2-[[(2S)-2-[[(2R)-2-[[2-[[2-[[2-[(2-amino-1-hydroxyethylidene)amino]-3-carboxy-1-hydroxypropylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxybutylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1,5-dihydroxy-5-iminopentylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxybutylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxyethylidene]amino]hexanoic acid Chemical compound C[C@@H]([C@@H](C(=N[C@@H](CS)C(=N[C@@H](C)C(=N[C@@H](CO)C(=NCC(=N[C@@H](CCC(=N)O)C(=NC(CS)C(=N[C@H]([C@H](C)O)C(=N[C@H](CS)C(=N[C@H](CO)C(=NCC(=N[C@H](CS)C(=NCC(=N[C@H](CCCCN)C(=O)O)O)O)O)O)O)O)O)O)O)O)O)O)O)N=C([C@H](CS)N=C([C@H](CO)N=C([C@H](CO)N=C([C@H](C)N=C(CN=C([C@H](CO)N=C([C@H](CS)N=C(CN=C(C(CS)N=C(C(CC(=O)O)N=C(CN)O)O)O)O)O)O)O)O)O)O)O)O DIGQNXIGRZPYDK-WKSCXVIASA-N 0.000 description 1
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 1
- XSAKVDNHFRWJKS-IIZANFQQSA-N (2s)-n-benzyl-1-[(2s)-1-[(2s)-2-[[(2s)-2-[[(2s)-2-(dimethylamino)-3-methylbutanoyl]amino]-3-methylbutanoyl]-methylamino]-3-methylbutanoyl]pyrrolidine-2-carbonyl]pyrrolidine-2-carboxamide Chemical compound CC(C)[C@H](N(C)C)C(=O)N[C@@H](C(C)C)C(=O)N(C)[C@@H](C(C)C)C(=O)N1CCC[C@H]1C(=O)N1[C@H](C(=O)NCC=2C=CC=CC=2)CCC1 XSAKVDNHFRWJKS-IIZANFQQSA-N 0.000 description 1
- MWWSFMDVAYGXBV-MYPASOLCSA-N (7r,9s)-7-[(2r,4s,5s,6s)-4-amino-5-hydroxy-6-methyloxan-2-yl]oxy-6,9,11-trihydroxy-9-(2-hydroxyacetyl)-4-methoxy-8,10-dihydro-7h-tetracene-5,12-dione;hydrochloride Chemical compound Cl.O([C@@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 MWWSFMDVAYGXBV-MYPASOLCSA-N 0.000 description 1
- IEXUMDBQLIVNHZ-YOUGDJEHSA-N (8s,11r,13r,14s,17s)-11-[4-(dimethylamino)phenyl]-17-hydroxy-17-(3-hydroxypropyl)-13-methyl-1,2,6,7,8,11,12,14,15,16-decahydrocyclopenta[a]phenanthren-3-one Chemical compound C1=CC(N(C)C)=CC=C1[C@@H]1C2=C3CCC(=O)C=C3CC[C@H]2[C@H](CC[C@]2(O)CCCO)[C@@]2(C)C1 IEXUMDBQLIVNHZ-YOUGDJEHSA-N 0.000 description 1
- LKJPYSCBVHEWIU-KRWDZBQOSA-N (R)-bicalutamide Chemical compound C([C@@](O)(C)C(=O)NC=1C=C(C(C#N)=CC=1)C(F)(F)F)S(=O)(=O)C1=CC=C(F)C=C1 LKJPYSCBVHEWIU-KRWDZBQOSA-N 0.000 description 1
- UKAUYVFTDYCKQA-UHFFFAOYSA-N -2-Amino-4-hydroxybutanoic acid Natural products OC(=O)C(N)CCO UKAUYVFTDYCKQA-UHFFFAOYSA-N 0.000 description 1
- TZCPCKNHXULUIY-RGULYWFUSA-N 1,2-distearoyl-sn-glycero-3-phosphoserine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP(O)(=O)OC[C@H](N)C(O)=O)OC(=O)CCCCCCCCCCCCCCCCC TZCPCKNHXULUIY-RGULYWFUSA-N 0.000 description 1
- ROZCIVXTLACYNY-UHFFFAOYSA-N 2,3,4,5,6-pentafluoro-n-(3-fluoro-4-methoxyphenyl)benzenesulfonamide Chemical compound C1=C(F)C(OC)=CC=C1NS(=O)(=O)C1=C(F)C(F)=C(F)C(F)=C1F ROZCIVXTLACYNY-UHFFFAOYSA-N 0.000 description 1
- BFSVOASYOCHEOV-UHFFFAOYSA-N 2-diethylaminoethanol Chemical compound CCN(CC)CCO BFSVOASYOCHEOV-UHFFFAOYSA-N 0.000 description 1
- YNFSUOFXEVCDTC-UHFFFAOYSA-N 2-n-methyl-7h-purine-2,6-diamine Chemical compound CNC1=NC(N)=C2NC=NC2=N1 YNFSUOFXEVCDTC-UHFFFAOYSA-N 0.000 description 1
- 108020005065 3' Flanking Region Proteins 0.000 description 1
- FFRFGVHNKJYNOV-DOVUUNBWSA-N 3',4'-Anhydrovinblastine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](OC(C)=O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(=O)OC)N3C)C=2)OC)C=C(C2)CC)N2CCC2=C1NC1=CC=CC=C21 FFRFGVHNKJYNOV-DOVUUNBWSA-N 0.000 description 1
- AZKSAVLVSZKNRD-UHFFFAOYSA-M 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide Chemical compound [Br-].S1C(C)=C(C)N=C1[N+]1=NC(C=2C=CC=CC=2)=NN1C1=CC=CC=C1 AZKSAVLVSZKNRD-UHFFFAOYSA-M 0.000 description 1
- 108020005029 5' Flanking Region Proteins 0.000 description 1
- WOVKYSAHUYNSMH-RRKCRQDMSA-N 5-bromodeoxyuridine Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(Br)=C1 WOVKYSAHUYNSMH-RRKCRQDMSA-N 0.000 description 1
- STQGQHZAVUOBTE-UHFFFAOYSA-N 7-Cyan-hept-2t-en-4,6-diinsaeure Natural products C1=2C(O)=C3C(=O)C=4C(OC)=CC=CC=4C(=O)C3=C(O)C=2CC(O)(C(C)=O)CC1OC1CC(N)C(O)C(C)O1 STQGQHZAVUOBTE-UHFFFAOYSA-N 0.000 description 1
- KABRXLINDSPGDF-UHFFFAOYSA-N 7-bromoisoquinoline Chemical compound C1=CN=CC2=CC(Br)=CC=C21 KABRXLINDSPGDF-UHFFFAOYSA-N 0.000 description 1
- MSSXOMSJDRHRMC-UHFFFAOYSA-N 9H-purine-2,6-diamine Chemical compound NC1=NC(N)=C2NC=NC2=N1 MSSXOMSJDRHRMC-UHFFFAOYSA-N 0.000 description 1
- 101800000263 Acidic protein Proteins 0.000 description 1
- HRPVXLWXLXDGHG-UHFFFAOYSA-N Acrylamide Chemical compound NC(=O)C=C HRPVXLWXLXDGHG-UHFFFAOYSA-N 0.000 description 1
- 206010000830 Acute leukaemia Diseases 0.000 description 1
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 description 1
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 description 1
- 206010000871 Acute monocytic leukaemia Diseases 0.000 description 1
- 208000031261 Acute myeloid leukaemia Diseases 0.000 description 1
- 206010000890 Acute myelomonocytic leukaemia Diseases 0.000 description 1
- 239000012110 Alexa Fluor 594 Substances 0.000 description 1
- 108091093088 Amplicon Proteins 0.000 description 1
- 201000003076 Angiosarcoma Diseases 0.000 description 1
- 108090000672 Annexin A5 Proteins 0.000 description 1
- 102000004121 Annexin A5 Human genes 0.000 description 1
- 108020005544 Antisense RNA Proteins 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 241000132092 Aster Species 0.000 description 1
- 206010003571 Astrocytoma Diseases 0.000 description 1
- 241000972773 Aulopiformes Species 0.000 description 1
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 description 1
- 208000032791 BCR-ABL1 positive chronic myelogenous leukemia Diseases 0.000 description 1
- 206010004146 Basal cell carcinoma Diseases 0.000 description 1
- 206010005003 Bladder cancer Diseases 0.000 description 1
- 108010006654 Bleomycin Proteins 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 241000701822 Bovine papillomavirus Species 0.000 description 1
- 238000009010 Bradford assay Methods 0.000 description 1
- 208000003174 Brain Neoplasms Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- 102100038451 CDK5 regulatory subunit-associated protein 2 Human genes 0.000 description 1
- RZZPDXZPRHQOCG-OJAKKHQRSA-O CDP-choline(1+) Chemical compound O[C@@H]1[C@H](O)[C@@H](COP(O)(=O)OP(O)(=O)OCC[N+](C)(C)C)O[C@H]1N1C(=O)N=C(N)C=C1 RZZPDXZPRHQOCG-OJAKKHQRSA-O 0.000 description 1
- QCMYYKRYFNMIEC-UHFFFAOYSA-N COP(O)=O Chemical class COP(O)=O QCMYYKRYFNMIEC-UHFFFAOYSA-N 0.000 description 1
- 241000244203 Caenorhabditis elegans Species 0.000 description 1
- 101100400999 Caenorhabditis elegans mel-28 gene Proteins 0.000 description 1
- 101710096866 Calmodulin-related protein Proteins 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 108090000397 Caspase 3 Proteins 0.000 description 1
- 102100029855 Caspase-3 Human genes 0.000 description 1
- 102000047934 Caspase-3/7 Human genes 0.000 description 1
- 108700037887 Caspase-3/7 Proteins 0.000 description 1
- 102000011727 Caspases Human genes 0.000 description 1
- 108010076667 Caspases Proteins 0.000 description 1
- 102100023441 Centromere protein J Human genes 0.000 description 1
- 206010008342 Cervix carcinoma Diseases 0.000 description 1
- 241000867607 Chlorocebus sabaeus Species 0.000 description 1
- 208000005243 Chondrosarcoma Diseases 0.000 description 1
- 201000009047 Chordoma Diseases 0.000 description 1
- 208000006332 Choriocarcinoma Diseases 0.000 description 1
- 208000010833 Chronic myeloid leukaemia Diseases 0.000 description 1
- 206010009944 Colon cancer Diseases 0.000 description 1
- 108020004635 Complementary DNA Proteins 0.000 description 1
- 108020004394 Complementary RNA Proteins 0.000 description 1
- 208000009798 Craniopharyngioma Diseases 0.000 description 1
- 229930188224 Cryptophycin Natural products 0.000 description 1
- 102100021906 Cyclin-O Human genes 0.000 description 1
- UHDGCWIWMRVCDJ-CCXZUQQUSA-N Cytarabine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 UHDGCWIWMRVCDJ-CCXZUQQUSA-N 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- 108010008286 DNA nucleotidylexotransferase Proteins 0.000 description 1
- 102100033215 DNA nucleotidylexotransferase Human genes 0.000 description 1
- 108010092160 Dactinomycin Proteins 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- LQKSHSFQQRCAFW-UHFFFAOYSA-N Dolastatin 15 Natural products COC1=CC(=O)N(C(=O)C(OC(=O)C2N(CCC2)C(=O)C2N(CCC2)C(=O)C(C(C)C)N(C)C(=O)C(NC(=O)C(C(C)C)N(C)C)C(C)C)C(C)C)C1CC1=CC=CC=C1 LQKSHSFQQRCAFW-UHFFFAOYSA-N 0.000 description 1
- 201000009051 Embryonal Carcinoma Diseases 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 206010014733 Endometrial cancer Diseases 0.000 description 1
- 206010014967 Ependymoma Diseases 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 208000031637 Erythroblastic Acute Leukemia Diseases 0.000 description 1
- 208000036566 Erythroleukaemia Diseases 0.000 description 1
- 241000206602 Eukaryota Species 0.000 description 1
- 208000006168 Ewing Sarcoma Diseases 0.000 description 1
- 108010074860 Factor Xa Proteins 0.000 description 1
- 241000282324 Felis Species 0.000 description 1
- 201000008808 Fibrosarcoma Diseases 0.000 description 1
- 241000700662 Fowlpox virus Species 0.000 description 1
- 101150066516 GST gene Proteins 0.000 description 1
- 208000032612 Glial tumor Diseases 0.000 description 1
- 206010018338 Glioma Diseases 0.000 description 1
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 1
- ZWZWYGMENQVNFU-UHFFFAOYSA-N Glycerophosphorylserin Natural products OC(=O)C(N)COP(O)(=O)OCC(O)CO ZWZWYGMENQVNFU-UHFFFAOYSA-N 0.000 description 1
- 241000282575 Gorilla Species 0.000 description 1
- 208000001258 Hemangiosarcoma Diseases 0.000 description 1
- 101100163968 Homo sapiens ASPM gene Proteins 0.000 description 1
- 101000756632 Homo sapiens Actin, cytoplasmic 1 Proteins 0.000 description 1
- 101000882873 Homo sapiens CDK5 regulatory subunit-associated protein 2 Proteins 0.000 description 1
- 101000897441 Homo sapiens Cyclin-O Proteins 0.000 description 1
- 241000701024 Human betaherpesvirus 5 Species 0.000 description 1
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 1
- PMMYEEVYMWASQN-DMTCNVIQSA-N Hydroxyproline Chemical compound O[C@H]1CN[C@H](C(O)=O)C1 PMMYEEVYMWASQN-DMTCNVIQSA-N 0.000 description 1
- VSNHCAURESNICA-UHFFFAOYSA-N Hydroxyurea Chemical compound NC(=O)NO VSNHCAURESNICA-UHFFFAOYSA-N 0.000 description 1
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 1
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 1
- 108010091135 Immunoglobulin Fc Fragments Proteins 0.000 description 1
- 102000018071 Immunoglobulin Fc Fragments Human genes 0.000 description 1
- 208000026350 Inborn Genetic disease Diseases 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- 150000008575 L-amino acids Chemical class 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- UKAUYVFTDYCKQA-VKHMYHEASA-N L-homoserine Chemical compound OC(=O)[C@@H](N)CCO UKAUYVFTDYCKQA-VKHMYHEASA-N 0.000 description 1
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- QEFRNWWLZKMPFJ-ZXPFJRLXSA-N L-methionine (R)-S-oxide Chemical compound C[S@@](=O)CC[C@H]([NH3+])C([O-])=O QEFRNWWLZKMPFJ-ZXPFJRLXSA-N 0.000 description 1
- QEFRNWWLZKMPFJ-UHFFFAOYSA-N L-methionine sulphoxide Natural products CS(=O)CCC(N)C(O)=O QEFRNWWLZKMPFJ-UHFFFAOYSA-N 0.000 description 1
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 1
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 1
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 1
- 102100020870 La-related protein 6 Human genes 0.000 description 1
- 108050008265 La-related protein 6 Proteins 0.000 description 1
- JVTAAEKCZFNVCJ-UHFFFAOYSA-M Lactate Chemical compound CC(O)C([O-])=O JVTAAEKCZFNVCJ-UHFFFAOYSA-M 0.000 description 1
- 208000018142 Leiomyosarcoma Diseases 0.000 description 1
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 1
- 108060001084 Luciferase Proteins 0.000 description 1
- 239000005089 Luciferase Substances 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 208000030289 Lymphoproliferative disease Diseases 0.000 description 1
- 102000043136 MAP kinase family Human genes 0.000 description 1
- 108091054455 MAP kinase family Proteins 0.000 description 1
- 241000282560 Macaca mulatta Species 0.000 description 1
- 208000007054 Medullary Carcinoma Diseases 0.000 description 1
- 208000000172 Medulloblastoma Diseases 0.000 description 1
- 206010027406 Mesothelioma Diseases 0.000 description 1
- 102000003792 Metallothionein Human genes 0.000 description 1
- 108090000157 Metallothionein Proteins 0.000 description 1
- 102000002151 Microfilament Proteins Human genes 0.000 description 1
- 108010040897 Microfilament Proteins Proteins 0.000 description 1
- 229930192392 Mitomycin Natural products 0.000 description 1
- 208000035489 Monocytic Acute Leukemia Diseases 0.000 description 1
- 108090000143 Mouse Proteins Proteins 0.000 description 1
- 102000007474 Multiprotein Complexes Human genes 0.000 description 1
- 108010085220 Multiprotein Complexes Proteins 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- 208000033761 Myelogenous Chronic BCR-ABL Positive Leukemia Diseases 0.000 description 1
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 1
- 208000033835 Myelomonocytic Acute Leukemia Diseases 0.000 description 1
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 1
- ZDZOTLJHXYCWBA-VCVYQWHSSA-N N-debenzoyl-N-(tert-butoxycarbonyl)-10-deacetyltaxol Chemical compound O([C@H]1[C@H]2[C@@](C([C@H](O)C3=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=4C=CC=CC=4)C[C@]1(O)C3(C)C)=O)(C)[C@@H](O)C[C@H]1OC[C@]12OC(=O)C)C(=O)C1=CC=CC=C1 ZDZOTLJHXYCWBA-VCVYQWHSSA-N 0.000 description 1
- 206010029260 Neuroblastoma Diseases 0.000 description 1
- 239000000020 Nitrocellulose Substances 0.000 description 1
- 238000000636 Northern blotting Methods 0.000 description 1
- 101710163270 Nuclease Proteins 0.000 description 1
- 108091005461 Nucleic proteins Proteins 0.000 description 1
- 239000004677 Nylon Substances 0.000 description 1
- 229910004679 ONO2 Inorganic materials 0.000 description 1
- 108020005187 Oligonucleotide Probes Proteins 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 241000282577 Pan troglodytes Species 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- 229930040373 Paraformaldehyde Natural products 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 102000035195 Peptidases Human genes 0.000 description 1
- 108010067902 Peptide Library Proteins 0.000 description 1
- 208000007641 Pinealoma Diseases 0.000 description 1
- 239000004793 Polystyrene Substances 0.000 description 1
- 239000004237 Ponceau 6R Substances 0.000 description 1
- 241000282405 Pongo abelii Species 0.000 description 1
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 description 1
- HFVNWDWLWUCIHC-GUPDPFMOSA-N Prednimustine Chemical compound O=C([C@@]1(O)CC[C@H]2[C@H]3[C@@H]([C@]4(C=CC(=O)C=C4CC3)C)[C@@H](O)C[C@@]21C)COC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 HFVNWDWLWUCIHC-GUPDPFMOSA-N 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 108010007568 Protamines Proteins 0.000 description 1
- 102000007327 Protamines Human genes 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 108010026552 Proteome Proteins 0.000 description 1
- 108091030071 RNAI Proteins 0.000 description 1
- 108090000244 Rat Proteins Proteins 0.000 description 1
- 208000006265 Renal cell carcinoma Diseases 0.000 description 1
- 201000000582 Retinoblastoma Diseases 0.000 description 1
- OWPCHSCAPHNHAV-UHFFFAOYSA-N Rhizoxin Natural products C1C(O)C2(C)OC2C=CC(C)C(OC(=O)C2)CC2CC2OC2C(=O)OC1C(C)C(OC)C(C)=CC=CC(C)=CC1=COC(C)=N1 OWPCHSCAPHNHAV-UHFFFAOYSA-N 0.000 description 1
- 108091028664 Ribonucleotide Proteins 0.000 description 1
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 1
- 230000018199 S phase Effects 0.000 description 1
- 239000012722 SDS sample buffer Substances 0.000 description 1
- 206010039491 Sarcoma Diseases 0.000 description 1
- 101710184528 Scaffolding protein Proteins 0.000 description 1
- 241000242677 Schistosoma japonicum Species 0.000 description 1
- 241000239226 Scorpiones Species 0.000 description 1
- 201000010208 Seminoma Diseases 0.000 description 1
- 229920002684 Sepharose Polymers 0.000 description 1
- 238000012300 Sequence Analysis Methods 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 208000005718 Stomach Neoplasms Diseases 0.000 description 1
- 101100054666 Streptomyces halstedii sch3 gene Proteins 0.000 description 1
- 241000701093 Suid alphaherpesvirus 1 Species 0.000 description 1
- 239000012505 Superdex™ Substances 0.000 description 1
- NAVMQTYZDKMPEU-UHFFFAOYSA-N Targretin Chemical compound CC1=CC(C(CCC2(C)C)(C)C)=C2C=C1C(=C)C1=CC=C(C(O)=O)C=C1 NAVMQTYZDKMPEU-UHFFFAOYSA-N 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-N Thiophosphoric acid Chemical class OP(O)(S)=O RYYWUUFWQRZTIU-UHFFFAOYSA-N 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 108090000190 Thrombin Proteins 0.000 description 1
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 1
- 208000024770 Thyroid neoplasm Diseases 0.000 description 1
- 101710120037 Toxin CcdB Proteins 0.000 description 1
- 101710195626 Transcriptional activator protein Proteins 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 1
- 102100040247 Tumor necrosis factor Human genes 0.000 description 1
- 108050002568 Tumor necrosis factor ligand superfamily member 6 Proteins 0.000 description 1
- 102100031988 Tumor necrosis factor ligand superfamily member 6 Human genes 0.000 description 1
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 description 1
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 1
- 238000005411 Van der Waals force Methods 0.000 description 1
- 208000014070 Vestibular schwannoma Diseases 0.000 description 1
- JXLYSJRDGCGARV-WWYNWVTFSA-N Vinblastine Natural products O=C(O[C@H]1[C@](O)(C(=O)OC)[C@@H]2N(C)c3c(cc(c(OC)c3)[C@]3(C(=O)OC)c4[nH]c5c(c4CCN4C[C@](O)(CC)C[C@H](C3)C4)cccc5)[C@@]32[C@H]2[C@@]1(CC)C=CCN2CC3)C JXLYSJRDGCGARV-WWYNWVTFSA-N 0.000 description 1
- 208000033559 Waldenström macroglobulinemia Diseases 0.000 description 1
- 208000008383 Wilms tumor Diseases 0.000 description 1
- KYIKRXIYLAGAKQ-UHFFFAOYSA-N abcn Chemical compound C1CCCCC1(C#N)N=NC1(C#N)CCCCC1 KYIKRXIYLAGAKQ-UHFFFAOYSA-N 0.000 description 1
- 229960004103 abiraterone acetate Drugs 0.000 description 1
- UVIQSJCZCSLXRZ-UBUQANBQSA-N abiraterone acetate Chemical compound C([C@@H]1[C@]2(C)CC[C@@H]3[C@@]4(C)CC[C@@H](CC4=CC[C@H]31)OC(=O)C)C=C2C1=CC=CN=C1 UVIQSJCZCSLXRZ-UBUQANBQSA-N 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 230000021736 acetylation Effects 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 208000004064 acoustic neuroma Diseases 0.000 description 1
- 208000017733 acquired polycythemia vera Diseases 0.000 description 1
- RJURFGZVJUQBHK-IIXSONLDSA-N actinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-IIXSONLDSA-N 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 208000021841 acute erythroid leukemia Diseases 0.000 description 1
- 208000011912 acute myelomonocytic leukemia M4 Diseases 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- 125000001931 aliphatic group Chemical group 0.000 description 1
- 125000002877 alkyl aryl group Chemical group 0.000 description 1
- SHGAZHPCJJPHSC-YCNIQYBTSA-N all-trans-retinoic acid Chemical compound OC(=O)\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C SHGAZHPCJJPHSC-YCNIQYBTSA-N 0.000 description 1
- VREFGVBLTWBCJP-UHFFFAOYSA-N alprazolam Chemical compound C12=CC(Cl)=CC=C2N2C(C)=NN=C2CN=C1C1=CC=CC=C1 VREFGVBLTWBCJP-UHFFFAOYSA-N 0.000 description 1
- 229960000473 altretamine Drugs 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 125000005122 aminoalkylamino group Chemical group 0.000 description 1
- BFNBIHQBYMNNAN-UHFFFAOYSA-N ammonium sulfate Chemical compound N.N.OS(O)(=O)=O BFNBIHQBYMNNAN-UHFFFAOYSA-N 0.000 description 1
- 229910052921 ammonium sulfate Inorganic materials 0.000 description 1
- 235000011130 ammonium sulphate Nutrition 0.000 description 1
- 239000003708 ampul Substances 0.000 description 1
- 208000036878 aneuploidy Diseases 0.000 description 1
- 231100001075 aneuploidy Toxicity 0.000 description 1
- 229950001104 anhydrovinblastine Drugs 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 125000000129 anionic group Chemical group 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 238000011091 antibody purification Methods 0.000 description 1
- 229940041181 antineoplastic drug Drugs 0.000 description 1
- 238000003491 array Methods 0.000 description 1
- 125000003710 aryl alkyl group Chemical group 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 108010044540 auristatin Proteins 0.000 description 1
- VSRXQHXAPYXROS-UHFFFAOYSA-N azanide;cyclobutane-1,1-dicarboxylic acid;platinum(2+) Chemical compound [NH2-].[NH2-].[Pt+2].OC(=O)C1(C(O)=O)CCC1 VSRXQHXAPYXROS-UHFFFAOYSA-N 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 150000001576 beta-amino acids Chemical class 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- 229960002938 bexarotene Drugs 0.000 description 1
- 229960000997 bicalutamide Drugs 0.000 description 1
- 230000000975 bioactive effect Effects 0.000 description 1
- 239000013060 biological fluid Substances 0.000 description 1
- 230000008827 biological function Effects 0.000 description 1
- 229960002685 biotin Drugs 0.000 description 1
- 235000020958 biotin Nutrition 0.000 description 1
- 239000011616 biotin Substances 0.000 description 1
- 201000001531 bladder carcinoma Diseases 0.000 description 1
- 229960001561 bleomycin Drugs 0.000 description 1
- OYVAGSVQBOHSSS-UAPAGMARSA-O bleomycin A2 Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCC[S+](C)C)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C OYVAGSVQBOHSSS-UAPAGMARSA-O 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 210000000601 blood cell Anatomy 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 201000010983 breast ductal carcinoma Diseases 0.000 description 1
- 208000003362 bronchogenic carcinoma Diseases 0.000 description 1
- 235000010633 broth Nutrition 0.000 description 1
- 229910001424 calcium ion Inorganic materials 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 238000007816 calorimetric assay Methods 0.000 description 1
- 125000000837 carbohydrate group Chemical group 0.000 description 1
- 229960004562 carboplatin Drugs 0.000 description 1
- UHBYWPGGCSDKFX-UHFFFAOYSA-N carboxyglutamic acid Chemical compound OC(=O)C(N)CC(C(O)=O)C(O)=O UHBYWPGGCSDKFX-UHFFFAOYSA-N 0.000 description 1
- 230000021523 carboxylation Effects 0.000 description 1
- 238000006473 carboxylation reaction Methods 0.000 description 1
- 229960005243 carmustine Drugs 0.000 description 1
- 230000003197 catalytic effect Effects 0.000 description 1
- 125000002091 cationic group Chemical group 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000022131 cell cycle Effects 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 229950009017 cemadotin Drugs 0.000 description 1
- 108010046713 cemadotin Proteins 0.000 description 1
- 210000003710 cerebral cortex Anatomy 0.000 description 1
- 201000010881 cervical cancer Diseases 0.000 description 1
- 238000012412 chemical coupling Methods 0.000 description 1
- 150000005829 chemical entities Chemical class 0.000 description 1
- 210000000038 chest Anatomy 0.000 description 1
- 229960004630 chlorambucil Drugs 0.000 description 1
- JCKYGMPEJWAADB-UHFFFAOYSA-N chlorambucil Chemical compound OC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 JCKYGMPEJWAADB-UHFFFAOYSA-N 0.000 description 1
- 230000010428 chromatin condensation Effects 0.000 description 1
- 239000013611 chromosomal DNA Substances 0.000 description 1
- 230000024321 chromosome segregation Effects 0.000 description 1
- 208000024207 chronic leukemia Diseases 0.000 description 1
- 208000032852 chronic lymphocytic leukemia Diseases 0.000 description 1
- 229960004316 cisplatin Drugs 0.000 description 1
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 230000024203 complement activation Effects 0.000 description 1
- 239000012141 concentrate Substances 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 239000013068 control sample Substances 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 238000000315 cryotherapy Methods 0.000 description 1
- 108010006226 cryptophycin Proteins 0.000 description 1
- PSNOPSMXOBPNNV-VVCTWANISA-N cryptophycin 1 Chemical compound C1=C(Cl)C(OC)=CC=C1C[C@@H]1C(=O)NC[C@@H](C)C(=O)O[C@@H](CC(C)C)C(=O)O[C@H]([C@H](C)[C@@H]2[C@H](O2)C=2C=CC=CC=2)C/C=C/C(=O)N1 PSNOPSMXOBPNNV-VVCTWANISA-N 0.000 description 1
- PSNOPSMXOBPNNV-UHFFFAOYSA-N cryptophycin-327 Natural products C1=C(Cl)C(OC)=CC=C1CC1C(=O)NCC(C)C(=O)OC(CC(C)C)C(=O)OC(C(C)C2C(O2)C=2C=CC=CC=2)CC=CC(=O)N1 PSNOPSMXOBPNNV-UHFFFAOYSA-N 0.000 description 1
- 125000000753 cycloalkyl group Chemical group 0.000 description 1
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 208000002445 cystadenocarcinoma Diseases 0.000 description 1
- 229960000684 cytarabine Drugs 0.000 description 1
- 230000002380 cytological effect Effects 0.000 description 1
- 230000009089 cytolysis Effects 0.000 description 1
- 229940127089 cytotoxic agent Drugs 0.000 description 1
- 238000002784 cytotoxicity assay Methods 0.000 description 1
- 231100000263 cytotoxicity test Toxicity 0.000 description 1
- 229960003901 dacarbazine Drugs 0.000 description 1
- 229960000640 dactinomycin Drugs 0.000 description 1
- STQGQHZAVUOBTE-VGBVRHCVSA-N daunorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 description 1
- 229960000975 daunorubicin Drugs 0.000 description 1
- 230000007547 defect Effects 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 238000001212 derivatisation Methods 0.000 description 1
- 239000003599 detergent Substances 0.000 description 1
- 238000010586 diagram Methods 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- PMMYEEVYMWASQN-UHFFFAOYSA-N dl-hydroxyproline Natural products OC1C[NH2+]C(C([O-])=O)C1 PMMYEEVYMWASQN-UHFFFAOYSA-N 0.000 description 1
- 229960003668 docetaxel Drugs 0.000 description 1
- AMRJKAQTDDKMCE-UHFFFAOYSA-N dolastatin Chemical compound CC(C)C(N(C)C)C(=O)NC(C(C)C)C(=O)N(C)C(C(C)C)C(OC)CC(=O)N1CCCC1C(OC)C(C)C(=O)NC(C=1SC=CN=1)CC1=CC=CC=C1 AMRJKAQTDDKMCE-UHFFFAOYSA-N 0.000 description 1
- 229930188854 dolastatin Natural products 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 238000010828 elution Methods 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 230000007515 enzymatic degradation Effects 0.000 description 1
- 208000037828 epithelial carcinoma Diseases 0.000 description 1
- JEFPWOBULVSOTM-PPHPATTJSA-N ethyl n-[(2s)-5-amino-2-methyl-3-phenyl-1,2-dihydropyrido[3,4-b]pyrazin-7-yl]carbamate;2-hydroxyethanesulfonic acid Chemical compound OCCS(O)(=O)=O.C=1([C@H](C)NC=2C=C(N=C(N)C=2N=1)NC(=O)OCC)C1=CC=CC=C1 JEFPWOBULVSOTM-PPHPATTJSA-N 0.000 description 1
- 229960005420 etoposide Drugs 0.000 description 1
- VJJPUSNTGOMMGY-MRVIYFEKSA-N etoposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@H](C)OC[C@H]4O3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 VJJPUSNTGOMMGY-MRVIYFEKSA-N 0.000 description 1
- 230000007717 exclusion Effects 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 235000013861 fat-free Nutrition 0.000 description 1
- 210000004996 female reproductive system Anatomy 0.000 description 1
- 238000000855 fermentation Methods 0.000 description 1
- 230000004151 fermentation Effects 0.000 description 1
- 210000000604 fetal stem cell Anatomy 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- DBEPLOCGEIEOCV-WSBQPABSSA-N finasteride Chemical compound N([C@@H]1CC2)C(=O)C=C[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H](C(=O)NC(C)(C)C)[C@@]2(C)CC1 DBEPLOCGEIEOCV-WSBQPABSSA-N 0.000 description 1
- 229960004039 finasteride Drugs 0.000 description 1
- MKXKFYHWDHIYRV-UHFFFAOYSA-N flutamide Chemical compound CC(C)C(=O)NC1=CC=C([N+]([O-])=O)C(C(F)(F)F)=C1 MKXKFYHWDHIYRV-UHFFFAOYSA-N 0.000 description 1
- 229960002074 flutamide Drugs 0.000 description 1
- 239000011888 foil Substances 0.000 description 1
- 231100000221 frame shift mutation induction Toxicity 0.000 description 1
- 238000002825 functional assay Methods 0.000 description 1
- 230000002538 fungal effect Effects 0.000 description 1
- 230000005021 gait Effects 0.000 description 1
- 206010017758 gastric cancer Diseases 0.000 description 1
- 238000002523 gelfiltration Methods 0.000 description 1
- 238000003633 gene expression assay Methods 0.000 description 1
- 230000009368 gene silencing by RNA Effects 0.000 description 1
- 208000016361 genetic disease Diseases 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 208000025750 heavy chain disease Diseases 0.000 description 1
- 201000002222 hemangioblastoma Diseases 0.000 description 1
- 125000000592 heterocycloalkyl group Chemical group 0.000 description 1
- UUVWYPNAQBNQJQ-UHFFFAOYSA-N hexamethylmelamine Chemical compound CN(C)C1=NC(N(C)C)=NC(N(C)C)=N1 UUVWYPNAQBNQJQ-UHFFFAOYSA-N 0.000 description 1
- 238000002744 homologous recombination Methods 0.000 description 1
- 230000006801 homologous recombination Effects 0.000 description 1
- 238000001794 hormone therapy Methods 0.000 description 1
- 150000002431 hydrogen Chemical group 0.000 description 1
- 229960001330 hydroxycarbamide Drugs 0.000 description 1
- 229910052588 hydroxylapatite Inorganic materials 0.000 description 1
- 229960002591 hydroxyproline Drugs 0.000 description 1
- 229960001101 ifosfamide Drugs 0.000 description 1
- HOMGKSMUEGBAAB-UHFFFAOYSA-N ifosfamide Chemical compound ClCCNP1(=O)OCCCN1CCCl HOMGKSMUEGBAAB-UHFFFAOYSA-N 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 230000000984 immunochemical effect Effects 0.000 description 1
- 230000009851 immunogenic response Effects 0.000 description 1
- 230000016784 immunoglobulin production Effects 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 238000001114 immunoprecipitation Methods 0.000 description 1
- 238000012744 immunostaining Methods 0.000 description 1
- 230000006057 immunotolerant effect Effects 0.000 description 1
- 239000007943 implant Substances 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 239000003978 infusion fluid Substances 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 239000000138 intercalating agent Substances 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 238000000185 intracerebroventricular administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 238000004255 ion exchange chromatography Methods 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- BPHPUYQFMNQIOC-NXRLNHOXSA-N isopropyl beta-D-thiogalactopyranoside Chemical compound CC(C)S[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O BPHPUYQFMNQIOC-NXRLNHOXSA-N 0.000 description 1
- 108010045069 keyhole-limpet hemocyanin Proteins 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 238000011005 laboratory method Methods 0.000 description 1
- 229950007056 liarozole Drugs 0.000 description 1
- UGFHIPBXIWJXNA-UHFFFAOYSA-N liarozole Chemical compound ClC1=CC=CC(C(C=2C=C3NC=NC3=CC=2)N2C=NC=C2)=C1 UGFHIPBXIWJXNA-UHFFFAOYSA-N 0.000 description 1
- 125000003473 lipid group Chemical group 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 206010024627 liposarcoma Diseases 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 229960002247 lomustine Drugs 0.000 description 1
- 229960003538 lonidamine Drugs 0.000 description 1
- WDRYRZXSPDWGEB-UHFFFAOYSA-N lonidamine Chemical compound C12=CC=CC=C2C(C(=O)O)=NN1CC1=CC=C(Cl)C=C1Cl WDRYRZXSPDWGEB-UHFFFAOYSA-N 0.000 description 1
- 238000004020 luminiscence type Methods 0.000 description 1
- 201000005296 lung carcinoma Diseases 0.000 description 1
- 208000037829 lymphangioendotheliosarcoma Diseases 0.000 description 1
- 208000012804 lymphangiosarcoma Diseases 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 230000003211 malignant effect Effects 0.000 description 1
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 230000010534 mechanism of action Effects 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 208000023356 medullary thyroid gland carcinoma Diseases 0.000 description 1
- 210000002780 melanosome Anatomy 0.000 description 1
- 229960001924 melphalan Drugs 0.000 description 1
- SGDBTWWWUNNDEQ-LBPRGKRZSA-N melphalan Chemical compound OC(=O)[C@@H](N)CC1=CC=C(N(CCCl)CCCl)C=C1 SGDBTWWWUNNDEQ-LBPRGKRZSA-N 0.000 description 1
- 206010027191 meningioma Diseases 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- MYWUZJCMWCOHBA-VIFPVBQESA-N methamphetamine Chemical compound CN[C@@H](C)CC1=CC=CC=C1 MYWUZJCMWCOHBA-VIFPVBQESA-N 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- 125000000250 methylamino group Chemical group [H]N(*)C([H])([H])[H] 0.000 description 1
- LSDPWZHWYPCBBB-UHFFFAOYSA-O methylsulfide anion Chemical compound [SH2+]C LSDPWZHWYPCBBB-UHFFFAOYSA-O 0.000 description 1
- 238000010208 microarray analysis Methods 0.000 description 1
- 210000003632 microfilament Anatomy 0.000 description 1
- 238000000520 microinjection Methods 0.000 description 1
- 230000029115 microtubule polymerization Effects 0.000 description 1
- 210000003879 microtubule-organizing center Anatomy 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 210000004080 milk Anatomy 0.000 description 1
- 210000001700 mitochondrial membrane Anatomy 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 230000026211 mitotic metaphase Effects 0.000 description 1
- 108091005601 modified peptides Proteins 0.000 description 1
- 125000004573 morpholin-4-yl group Chemical group N1(CCOCC1)* 0.000 description 1
- 230000004660 morphological change Effects 0.000 description 1
- 239000012120 mounting media Substances 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 210000001167 myeloblast Anatomy 0.000 description 1
- 208000001611 myxosarcoma Diseases 0.000 description 1
- ZIUHHBKFKCYYJD-UHFFFAOYSA-N n,n'-methylenebisacrylamide Chemical compound C=CC(=O)NCNC(=O)C=C ZIUHHBKFKCYYJD-UHFFFAOYSA-N 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 208000007538 neurilemmoma Diseases 0.000 description 1
- 230000001272 neurogenic effect Effects 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- XWXYUMMDTVBTOU-UHFFFAOYSA-N nilutamide Chemical compound O=C1C(C)(C)NC(=O)N1C1=CC=C([N+]([O-])=O)C(C(F)(F)F)=C1 XWXYUMMDTVBTOU-UHFFFAOYSA-N 0.000 description 1
- 229960002653 nilutamide Drugs 0.000 description 1
- 229920001220 nitrocellulos Polymers 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 125000001893 nitrooxy group Chemical group [O-][N+](=O)O* 0.000 description 1
- 229920001778 nylon Polymers 0.000 description 1
- 239000002751 oligonucleotide probe Substances 0.000 description 1
- 238000002515 oligonucleotide synthesis Methods 0.000 description 1
- 229950011093 onapristone Drugs 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 125000001181 organosilyl group Chemical group [SiH3]* 0.000 description 1
- 230000003204 osmotic effect Effects 0.000 description 1
- 201000008968 osteosarcoma Diseases 0.000 description 1
- 201000002528 pancreatic cancer Diseases 0.000 description 1
- 208000008443 pancreatic carcinoma Diseases 0.000 description 1
- 208000004019 papillary adenocarcinoma Diseases 0.000 description 1
- 201000010198 papillary carcinoma Diseases 0.000 description 1
- 229920002866 paraformaldehyde Polymers 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- XYJRXVWERLGGKC-UHFFFAOYSA-D pentacalcium;hydroxide;triphosphate Chemical compound [OH-].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O XYJRXVWERLGGKC-UHFFFAOYSA-D 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 238000010647 peptide synthesis reaction Methods 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 230000002085 persistent effect Effects 0.000 description 1
- 230000003094 perturbing effect Effects 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 230000003285 pharmacodynamic effect Effects 0.000 description 1
- 239000012071 phase Substances 0.000 description 1
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 150000004713 phosphodiesters Chemical group 0.000 description 1
- BZQFBWGGLXLEPQ-REOHCLBHSA-N phosphoserine Chemical compound OC(=O)[C@@H](N)COP(O)(O)=O BZQFBWGGLXLEPQ-REOHCLBHSA-N 0.000 description 1
- USRGIUJOYOXOQJ-GBXIJSLDSA-N phosphothreonine Chemical compound OP(=O)(O)O[C@H](C)[C@H](N)C(O)=O USRGIUJOYOXOQJ-GBXIJSLDSA-N 0.000 description 1
- 238000005375 photometry Methods 0.000 description 1
- 208000024724 pineal body neoplasm Diseases 0.000 description 1
- 201000004123 pineal gland cancer Diseases 0.000 description 1
- 210000002381 plasma Anatomy 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 229920000729 poly(L-lysine) polymer Polymers 0.000 description 1
- 208000037244 polycythemia vera Diseases 0.000 description 1
- 229920000656 polylysine Polymers 0.000 description 1
- 229920002223 polystyrene Polymers 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 229960004694 prednimustine Drugs 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 230000037452 priming Effects 0.000 description 1
- CPTBDICYNRMXFX-UHFFFAOYSA-N procarbazine Chemical compound CNNCC1=CC=C(C(=O)NC(C)C)C=C1 CPTBDICYNRMXFX-UHFFFAOYSA-N 0.000 description 1
- 229960000624 procarbazine Drugs 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 210000004765 promyelocyte Anatomy 0.000 description 1
- 230000031877 prophase Effects 0.000 description 1
- 229940048914 protamine Drugs 0.000 description 1
- 230000006916 protein interaction Effects 0.000 description 1
- 230000007026 protein scission Effects 0.000 description 1
- 210000001938 protoplast Anatomy 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 238000002708 random mutagenesis Methods 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 230000008844 regulatory mechanism Effects 0.000 description 1
- 230000003362 replicative effect Effects 0.000 description 1
- 125000006853 reporter group Chemical group 0.000 description 1
- 239000011347 resin Substances 0.000 description 1
- 229920005989 resin Polymers 0.000 description 1
- 108091008146 restriction endonucleases Proteins 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 201000009410 rhabdomyosarcoma Diseases 0.000 description 1
- OWPCHSCAPHNHAV-LMONGJCWSA-N rhizoxin Chemical compound C/C([C@H](OC)[C@@H](C)[C@@H]1C[C@H](O)[C@]2(C)O[C@@H]2/C=C/[C@@H](C)[C@]2([H])OC(=O)C[C@@](C2)(C[C@@H]2O[C@H]2C(=O)O1)[H])=C\C=C\C(\C)=C\C1=COC(C)=N1 OWPCHSCAPHNHAV-LMONGJCWSA-N 0.000 description 1
- 239000002336 ribonucleotide Substances 0.000 description 1
- 125000002652 ribonucleotide group Chemical group 0.000 description 1
- 229920002477 rna polymer Polymers 0.000 description 1
- 235000019515 salmon Nutrition 0.000 description 1
- 206010039667 schwannoma Diseases 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 201000008407 sebaceous adenocarcinoma Diseases 0.000 description 1
- 210000000582 semen Anatomy 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 238000002864 sequence alignment Methods 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 238000002741 site-directed mutagenesis Methods 0.000 description 1
- 208000000587 small cell lung carcinoma Diseases 0.000 description 1
- 239000006104 solid solution Substances 0.000 description 1
- 210000001082 somatic cell Anatomy 0.000 description 1
- 206010041823 squamous cell carcinoma Diseases 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 239000008227 sterile water for injection Substances 0.000 description 1
- 201000011549 stomach cancer Diseases 0.000 description 1
- 229960001052 streptozocin Drugs 0.000 description 1
- ZSJLQEPLLKMAKR-GKHCUFPYSA-N streptozocin Chemical compound O=NN(C)C(=O)N[C@H]1[C@@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O ZSJLQEPLLKMAKR-GKHCUFPYSA-N 0.000 description 1
- 230000004960 subcellular localization Effects 0.000 description 1
- 125000001424 substituent group Chemical group 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 201000010965 sweat gland carcinoma Diseases 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 206010042863 synovial sarcoma Diseases 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 229960001603 tamoxifen Drugs 0.000 description 1
- 229960003102 tasonermin Drugs 0.000 description 1
- 230000016853 telophase Effects 0.000 description 1
- 210000001103 thalamus Anatomy 0.000 description 1
- 229960004072 thrombin Drugs 0.000 description 1
- 201000002510 thyroid cancer Diseases 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 238000006257 total synthesis reaction Methods 0.000 description 1
- FGMPLJWBKKVCDB-UHFFFAOYSA-N trans-L-hydroxy-proline Natural products ON1CCCC1C(O)=O FGMPLJWBKKVCDB-UHFFFAOYSA-N 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 230000002463 transducing effect Effects 0.000 description 1
- 238000003151 transfection method Methods 0.000 description 1
- 238000011426 transformation method Methods 0.000 description 1
- 238000002834 transmittance Methods 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 229960001727 tretinoin Drugs 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 125000000876 trifluoromethoxy group Chemical group FC(F)(F)O* 0.000 description 1
- 239000013638 trimer Substances 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 239000000107 tumor biomarker Substances 0.000 description 1
- 238000003160 two-hybrid assay Methods 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 241001515965 unidentified phage Species 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- 208000010570 urinary bladder carcinoma Diseases 0.000 description 1
- 208000012991 uterine carcinoma Diseases 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- 229960003048 vinblastine Drugs 0.000 description 1
- JXLYSJRDGCGARV-XQKSVPLYSA-N vincaleukoblastine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](OC(C)=O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(=O)OC)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1NC1=CC=CC=C21 JXLYSJRDGCGARV-XQKSVPLYSA-N 0.000 description 1
- 229960004528 vincristine Drugs 0.000 description 1
- OGWKCGZFUXNPDA-UHFFFAOYSA-N vincristine Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(OC(C)=O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-UHFFFAOYSA-N 0.000 description 1
- OGWKCGZFUXNPDA-XQKSVPLYSA-N vincristine Chemical compound C([N@]1C[C@@H](C[C@]2(C(=O)OC)C=3C(=CC4=C([C@]56[C@H]([C@@]([C@H](OC(C)=O)[C@]7(CC)C=CCN([C@H]67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)C[C@@](C1)(O)CC)CC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-XQKSVPLYSA-N 0.000 description 1
- 229960005212 vindesine sulfate Drugs 0.000 description 1
- 229960000922 vinflunine Drugs 0.000 description 1
- NMDYYWFGPIMTKO-HBVLKOHWSA-N vinflunine Chemical compound C([C@@](C1=C(C2=CC=CC=C2N1)C1)(C2=C(OC)C=C3N(C)[C@@H]4[C@@]5(C3=C2)CCN2CC=C[C@]([C@@H]52)([C@H]([C@]4(O)C(=O)OC)OC(C)=O)CC)C(=O)OC)[C@H]2C[C@@H](C(C)(F)F)CN1C2 NMDYYWFGPIMTKO-HBVLKOHWSA-N 0.000 description 1
- 239000008215 water for injection Substances 0.000 description 1
Classifications
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/574—Immunoassay; Biospecific binding assay; Materials therefor for cancer
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6876—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
- C12Q1/6883—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
- C12Q1/6886—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q2600/00—Oligonucleotides characterized by their use
- C12Q2600/136—Screening for pharmacological compounds
Definitions
- Gynecological cancers such as uterine carcinoma and ovarian cancer are critical problems for women.
- Uterine cancer is the most common cancer in the United States among women.
- Ovarian cancer ranks fifth in cancer deaths among women, totaling more deaths than any other cancer of the female reproductive system. This may reflect the lack of a diagnostic test capable of diagnosing ovarian cancer in its early stages when it is most amenable to treatment.
- the effective treatment of ovarian cancer will likely be significantly increased if a screening test to identify early stage ovarian cancer is developed. Given the high incidence of ovarian cancer and other neoplasias, there is an urgent need for new diagnostic and therapeutic methods.
- the present invention provides for diagnostic methods, compositions, and kits that are useful for identifying a neoplasia by measuring Abnormal SPindle-like Microcephaly associated (ASPM) expression in a patient sample.
- the invention provides for methods of treating a neoplasia having increased ASPM expression.
- the invention generally features a method of diagnosing a subject (e.g., a human) as having, or having a propensity to develop, a neoplasia (e.g., cancer of the brain, breast, lung, pancreas, uterus, colon, thyroid, liver, bladder, kidney, ovary, testis, stomach, lymph node, cervix, and esophagus).
- a neoplasia e.g., cancer of the brain, breast, lung, pancreas, uterus, colon, thyroid, liver, bladder, kidney, ovary, testis, stomach, lymph node, cervix, and esophagus.
- the method involves determining the level of expression of an ASPM nucleic acid molecule (e.g., any nucleic acid sequence provided herein) in a subject sample, where an increased level of expression relative to a reference, indicates that the subject has or has a propensity to develop a neoplasia
- the invention features a method of diagnosing a subject as having, or having a propensity to develop, a neoplasia.
- the method involves determining the level of expression of an ASPM polypeptide (e.g., any amino acid sequence provided herein) in a subject sample, where an increased level (e.g., increased by at least 5%, 10%, 25%, 50%, 75%, 85%, 90% or 95%; or where the increase is by 5X, 10X, 2OX or 30X) of expression relative to the level of expression in a reference, indicates that the subject has or has a propensity to develop a neoplasia.
- the method involves determining the level of expression of the ASPM polypeptide (e.g., using an immunological assay).
- the invention features a method of diagnosing a subject as having, or having a propensity to develop, a neoplasia.
- the method involves determining the level of biological activity (e.g., mitotic spindle activity, calmodulin binding activity, cell proliferative active, maintenance of chromosomal stability) of an ASPM polypeptide in a subject sample, where an alteration in the level of biological activity relative to the biological activity in a reference, indicates that the subject has or has a propensity to develop a neoplasia.
- biological activity e.g., mitotic spindle activity, calmodulin binding activity, cell proliferative active, maintenance of chromosomal stability
- the invention features a method of monitoring a subject diagnosed as having a neoplasia.
- the method involves determining the level of an ASPM polypeptide in a subject sample, where an alteration in the level of expression relative to the level of expression in a reference indicates the severity of neoplasia in the subject.
- the subject is being treated for a neoplasia (e.g. breast, prostate, lung, testis, ovary, or uterine neoplasia).
- the alteration is an increase or a decrease.
- the increase indicates an increased severity of neoplasia.
- the reference is a control subject sample (e.g., a biological sample, such as a biological fluid or tissue sample).
- the reference is a sample obtained at an earlier time point.
- the method is used to diagnose a subject as having neoplasia; is used to determine the treatment regimen for a subject having neoplasia; is used to monitor the condition of a subject being treated for neoplasia; or is used to determine the prognosis of a subject having neoplasia.
- a poor prognosis determines an aggressive treatment regimen for the subject.
- the invention provides a method for identifying a subject as having or having a propensity to develop a neoplasia.
- the method involves detecting an alteration in the sequence of an ASPM nucleic acid molecule relative to the sequence or expression of a reference molecule.
- the alteration is detected using a hybridization reaction or is detected by sequencing the nucleic acid molecule.
- the invention provides an ASPM antibody that specifically binds to an ASPM protein or fragment thereof (e.g., a VTRK. or QSPE epitope of an ASPM polypeptide).
- the invention features a polypeptide containing an isolated ASPM protein variant, or fragment thereof, having substantial identity to ASPM variant 1, 2, or 3, where the polypeptide has an ASPM biological activity (e.g., mitotic spindle activity, calmodulin binding activity, functions in cell proliferation, or functions in chromosomal stability).
- ASPM protein variant is at least 80%, 85%, 90%, or 95% identical to ASPM variant 1, 2, or 3.
- ASPM protein variant contains at least an IQ domain and is capable of binding calmodulin.
- the polypeptide consists of an ASPM protein variant selected from the group consisting of ASPM variant 1, 2, and 3.
- the polypeptide is a fusion protein; is linked to a detectable amino acid sequence; or is linked to an affinity tag.
- the invention features an isolated ASPM nucleic acid molecule, where the nucleic acid molecule encodes a polypeptide of any of the previous aspects, a vector comprising such a nucleic acid molecule, and a host cell (e.g., in vitro or in vivo) comprising the vector or nucleic acid molecule.
- a host cell e.g., in vitro or in vivo
- the invention features an isolated ASPM inhibitory nucleic acid molecule, where the inhibitory nucleic acid molecule specifically binds at least a fragment of a nucleic acid molecule encoding an ASPM protein; a vector containing a nucleic acid molecule encoding the ASPM inhibitory nucleic acid molecule; and a host cell (e.g., in vitro or in vivo) comprising the vector or nucleic acid molecule.
- a host cell e.g., in vitro or in vivo
- the vector is an expression vector where the nucleic acid molecule is positioned for expression.
- the nucleic acid molecule is operably linked to a promoter (e.g., a promoter suitable for expression in a mammalian cell).
- the host cell expresses an ASPM protein variant.
- the host cell is a mammalian cell, such as a human cell.
- the invention features a double-stranded RNA corresponding to at least a portion of an ASPM nucleic acid molecule that encodes an ASPM protein, where the double-stranded RNA is capable of altering protein expression level.
- the double-stranded RNA is capable of altering protein expression level.
- RNA is an siRNA.
- the invention features an antisense nucleic acid molecule, where the antisense nucleic acid molecule is complementary to at least six nucleotides of an ASPM nucleic acid molecule that encodes an ASPM protein, and where the antisense is capable of altering expression from the nucleic acid molecule to which it is complementary.
- the invention features a primer capable of binding to an ASPM nucleic acid molecule encoding an ASPM protein variant that is upregulated in a neoplastic tissue.
- the invention features a collection of primers capable of binding to and amplifying an ASPM nucleic acid molecule, where at least one of the primers in the collection is the primer of the previous aspect.
- the collection features at least one pair of primers capable of amplifying an ASPM protein (e.g., ASPM, ASPM variant
- the primers are useful in diagnosing a neoplasia.
- the invention features a pharmaceutical composition containing an effective amount of an ASPM protein, variant, or fragment thereof, in a pharmaceutically acceptable excipient, where the fragment is capable of modulating (i.e., increasing or decreasing) cell proliferation or chromosome stability.
- the invention features a pharmaceutical composition containing an effective amount of a nucleic acid molecule of any previous aspect in a pharmaceutically acceptable excipient, where the fragment is capable of modulating cell proliferation or chromosomal stability.
- the invention features a pharmaceutical composition containing an effective amount of a vector containing a nucleic acid molecule encoding an ASPM protein of any previous aspect in a pharmaceutically acceptable excipient, where expression of the ASPM protein in a cell is capable of reducing, stabilizing, or inhibiting a neoplasia, decreasing cell proliferation or enhancing chromosomal stability.
- the vector containing a nucleic acid molecule encoding an ASPM protein of any previous aspect in a pharmaceutically acceptable excipient, where expression of the ASPM protein in a cell is capable of reducing, stabilizing, or inhibiting a neoplasia, decreasing cell proliferation or enhancing chromosomal stability.
- ASPM nucleic acid molecule is positioned for expression in a mammalian cell.
- the invention features an ASPM biomarker purified on a biochip.
- the invention features a microarray containing at least two nucleic acid molecules, or fragments thereof, fixed to a solid support, where at least one of the nucleic acid molecules is an ASPM nucleic acid molecule.
- the invention features a microarray containing at least two polypeptides, or fragments thereof, bound to a solid support, where at least one of the polypeptides on the support is an ASPM polypeptide.
- the invention features a diagnostic kit for the diagnosis of a neoplasia in a subject containing an ASPM nucleic acid molecule, or fragment thereof, and written instructions for use of the kit for detection of a neoplasia.
- the invention features a diagnostic kit for the diagnosis of a neoplasia in a subject containing an antibody that specifically binds an ASPM polypeptide, or fragment thereof, and written instructions for use of the kit for detection of a neoplasia.
- the invention features a kit identifying a subject as having or having a propensity to develop a neoplasia, containing an adsorbent, where the adsorbent retains an ASPM biomarker, and written instructions for use of the kit for detection of a neoplasia.
- the invention features a kit containing a first capture reagent that specifically binds an ASPM biomarker, and written instructions for use of the kit for detection of a neoplasia.
- the invention features a method for detecting neoplasia in a subject sample, where the method involves (a) contacting a subject sample with a capture reagent affixed to a substrate; and (b) capturing an ASPM polypeptide or nucleic acid molecule with the capture reagent.
- the subject sample comprises an ASPM protein or fragment thereof.
- the ASPM protein is fractionated prior to contacting the capture reagent.
- the invention features a method of altering the expression of an ASPM nucleic acid molecule in a cell, the method comprising contacting the cell with an effective amount of a compound capable of altering the expression of the ASPM nucleic acid molecule.
- the compound is an ASPM antisense nucleic acid molecule, a small interfering RNA (siRNA), or a double stranded RNA (dsRNA) that inhibits the expression of an ASPM nucleic acid molecule.
- the invention features a method of altering ASPM protein expression in a cell, the method comprising contacting the cell with a compound capable of altering the expression of an ASPM polypeptide.
- the cell is a human cell, a neoplastic cell, a cell is in vitro or a cell in vivo.
- the invention features a method of treating or preventing a neoplasia. The method involves administering to a subject (e.g., a human) in need thereof an effective amount of a pharmaceutical composition that alters expression of an ASPM polypeptide.
- the invention features a method of identifying a compound that inhibits a neoplasia.
- the method involves contacting a cell that expresses an ASPM nucleic acid molecule with a candidate compound, and comparing the level of expression of the nucleic acid molecule in the cell contacted by the candidate compound with the level of expression in a control cell not contacted by the candidate compound, where an alteration in expression of the ASPM nucleic acid molecule identifies the candidate compound as a compound that inhibits a neoplasia.
- the invention features a method of identifying a compound that inhibits a neoplasia.
- the method involves contacting a cell that expresses an ASPM polypeptide with a candidate compound, and comparing the level of expression of the polypeptide in the cell contacted by the candidate compound with the level of polypeptide expression in a control cell not contacted by the candidate compound, where an alteration in the expression of the ASPM polypeptide identifies the candidate compound as a compound that inhibits a neoplasia.
- the invention features a method of identifying a compound that inhibits a neoplasia.
- the method involves contacting a cell that expresses an ASPM polypeptide with a candidate compound, and comparing the biological activity of the polypeptide in the cell contacted by the candidate compound with the level of biological activity in a control cell not contacted by the candidate compound, where an alteration in the biological activity of the ASPM polypeptide identifies the candidate compound as a candidate compound that inhibits a neoplasia.
- the invention features method of identifying a candidate compound that inhibits a neoplasia, the method comprising a) contacting a cell containing an ASPM nucleic acid molecule present in an expression vector that includes a reporter construct; b) detecting the level of reporter gene expression in the cell contacted with the candidate compound with a control cell not contacted with the candidate compound, where an alteration in the level of the reporter gene expression identifies the candidate compound as a candidate compound that inhibits a neoplasia.
- the alteration in expression is an increase or a decrease in transcription. In other embodiments, the alteration in expression is an increase or a decrease in translation. In still other embodiments, the cell is in vitro or in vivo. In still other embodiments, the cell is a human cell, such as a neoplastic cell. In yet other embodiments, the alteration in expression is assayed using an immunological assay, an enzymatic assay, or a radioimmunoassay. In various embodiments of the above aspects, the cell is a human cell, a neoplastic cell, a cell is in vitro or a cell in vivo.
- ASPM protein or nucleic acid molecule expression is increased in a neoplastic cell by at least 2X, 5X, 10X, 2OX, 30X, 4OX, 5OX the level of expression in a corresponding control cell.
- a method or composition that alters (e.g., increases or decreases) ASPM expression by at least 5%, 10%, 25%, 50%, 75% or 100% is useful in the invention.
- Figures IA and IB show mouse and human ASPM expression in a variety of tissues.
- Figure IB is a photograph of an agarose gel showing relative levels of human ASPM expression in embryonic tissues. The ASPM transcripts in various tissues were analyzed using RT-PCR. Expression analyses of the mRNA was performed using human multiple fetal tissue cDNAs. ⁇ -actin was used as an internal control.
- Figures 2A and 2B are graphs showing expression of ASPM ia ovarian and uterine cancers.
- Figures 3 A and 3B show RT-PCR expression of ASPM in various human and mouse tissues.
- Figure 3A shows expression in human normal and matching tumor tissues.
- Figure 4B shows expression in normal murine tissues.
- RT-PCR expression of actin is provided as a reference.
- Figures 4A and 4B are photographs of an agarose gel containing alternatively spliced ASPM variants in fetal tissues (Figure 4A) and ⁇ -actin ( Figure 4B) visualized using ethidium bromide. Two major ASPM transcripts with sizes of ⁇ 10.3 and 5.7 kb were identified in all tissues analyzed. Additional spliced variants with variable sizes are also seen, ⁇ -actin was used as the internal control.
- Figures 5A and 5B show ASPM spliced forms in human and mouse.
- Figure 5A is a schematic diagram that shows the positions of major domains in the ASPM protein.
- the putative microtubule-binding domain is in gray, the calponin-homology domain in orange (positions 960-1056 and 1114-1174), IQ repeats in blue, and the terminal domain in black. Yellow color marks positions of newly identified 32 and 35 as long repeats in the N-terminus (see B).
- the middle part shows major spliced variants and comparison with the full-length ASPM protein.
- the bottom part shows parts of the human ASPM protein encoded by individual exons.
- Spliced variant 2 contains exon 26 followed by a part of intron 26 where the ORF is disrupted by stop codon. To better separate individual exons, the odd numbered exons are colored in black and even numbered ones in white.
- Figure 5B shows sequence alignments among mammalian species for two ASPM repeats identified herein (SEQ ID Nos: 1-16).
- the human ASPM protein contains two repeats in positions 316-347 and 366-400. Thee repeats are found in other mammals and one is also preserved in chicken. Identical amino acids are identified in black boxes. Residues shown in upper case font are amino acids that are invariant among the species in the alignment. Residues shown in lower case font are amino acids that are conserved, but that are not invariant. Gray shading identifies conservative substitutions.
- Figures 6A-6D show the structure and evolution of human ASPM IQ repeats (SEQ ID Nos: 1-16).
- the human ASPM protein contains two repeats in positions 316-347 and 366-400.
- Figure 6 A shows the organization of IQ repeats in the human ASPM protein. At the far left of Figure 6A, the number of IQ repeats and their positions in the full-length ASPM protein is shown. The length of the individual repeat is also indicated by amino acid residue numbers.
- the human alternatively spliced variants are highlighted in orange. Note that variants 2 and 3 contain large deletions that extend further toward the C-terminus of ASPM. The blue box displays the region deleted in mouse and rat.
- Color is used to mark positions that are variable in the analyzed primates (green monkey, rhesus monkey, orangutan, gorilla, chimpanzee, and human) and also changes specific to African hominoids. Based on the Gonnet PAM250 matrix, substitutions were divided into noncoservative (P ⁇ 0.5), and conservative (the rest).
- the IQ repeats 4-54 form an organized array of longer (-27 aa) and shorter ( ⁇ 23 aa) units. Alignment and conservation is shown separately for long ( Figure 6B) and short repeats ( Figure 6C) from the IQ4-54 region.
- Figure 6D shows the structure and evolution of mouse ASPM IQ repeats.
- the left column shows the number of IQ repeats and their positions in the full-length ASPM protein; the right column shows the length of the individual repeats.
- the grey positions mark sites that are different between mouse and rat proteins.
- Figures 7A-7I are photomicrographs showing the cytological analysis of ASPM proteins in mitotic cells HT1080 cells.
- HT1080 cells grown on coverslips were analyzed by indirect immunofluorescent staining using antibodies against human ASPM ( Figures 7B, 7E, and 7H). Chromosomal DNA was counterstained with DAPI ( Figures 7A, 7D and 7G).
- Anti-ASPM staining shows characteristic spindle pole staining patterns during mitotic periods, prometaphase (VTRK), metaphase (QSPE) and anaphase (VTRK). Scale bar indicates 10 ⁇ m.
- Figures 7 J, 7K, and 7L show a characterization of the anti-VTKR antibody.
- Figure 7A shows the analysis of protein extracts from a normal and a microcephalic individuals that were run on an SDS PAGE gel, transferred to a membrane, and immunoblotted with the anti- VTKR antibody.
- the Western blot in Figure 7 J shows the presence of two predicted ASPM proteins (a full-size 410 kDa protein and a 218 kDa isoform lacking an exon 18-encoded segment) in a protein extract derived from normal human cells (HT1080) (human lane).
- the predicted isoforms are 35 kDa shorter (i.e., 385 kDa and 183 kDa) in MM10458 cell protein extracts, which are derived from a microcephalic individual (patient lane).
- ASPM is truncated by a frameshift in exon 24 in MMl 0458.
- the anti-VTKR antibody also visualizes at least three additional bands in the HT 1080 extract; two of these are missing in the MMl 0458 extract. These bands may correspond to the predicted 164 kDa and 124 kDa ASPM isoforms.
- Figure 7K shows an immunoblot analysis of mouse ASPM proteins using the anti-VTKR antibody.
- the largest immunoreactive band in the mouse cell extract apparently corresponds to the predicted full-size 364 kDa ASPM protein that is 46 kDa shorter than human ASPM.
- the second major band apparently corresponds to the predicted 212 kDa mouse ASPM isoform lacking the exon 18-encoding segment that is approximately the same size as human ASPM isoform (218 kDa).
- Figure 7J i.e., ⁇ 150 kDa
- Figure 7L is a Western blot analysis showing the specificity of the affinity-purified anti-VTKR antibody.
- the antibody recognizes a recombinant MBP-ASPM fusion protein expressed in the pMAL-p2X expression vector.
- Protein extract 100 ⁇ g
- anti-VTKR antibody Lanes 1-4
- anti-MBP antibody Lanes 5- 8
- Lanes 1 and 5 correspond to the vector without insert.
- the predicted 60 kDa band corresponding to the fusion protein was detected.
- Lanes 2, 3, 4, 6, 7, and 8 correspond to individual transformants.
- Figures 8A-8J shows the sequences of the human ASPM polypeptide (Figure 8A), its variants 1 ( Figure 8C), 2 ( Figure 8E), and 3 ( Figure 8G), and the nucleic acid molecules encoding each of these polypeptides ( Figures 8B, 8D, 8F, 8H).
- Figures 81 and 8 J provide the nucleic acid and amino acid sequences of murine Aspm.
- ASPM polypeptide is meant a protein or protein variant, or fragment thereof, that is substantially identical to at least a portion of GenBank Accession No. NP__060606 (human ASPM) and that has an ASPM biological activity (e.g., calmodulin binding, function in mitosis, cell proliferation, regulation of symmetric cell division, or chromosomal stability).
- ASPM biological activity e.g., calmodulin binding, function in mitosis, cell proliferation, regulation of symmetric cell division, or chromosomal stability.
- ASPM nucleic acid molecule is meant a polynucleotide encoding an ASPM polypeptide or variant, or fragment thereof.
- APM biological activity is meant calmodulin binding activity, function during cell division, maintenance of chromosome stability, regulation of symmetric cell division, or any other function at a mitotic spindle, function in a neoplastic cell, or ASPM antibody binding.
- alteration is meant a change relative to a reference. For example, a change (increase or decrease) in the expression levels of a gene or polypeptide as detected by standard art known methods such as those described above.
- an alteration includes a 10% change in expression levels, preferably a 25% change, more preferably a 40% change, and most preferably a 50%, 75%, 85%, 95% or greater change in expression levels.
- biomarker is meant any protein or polynucleotide having an alteration in expression level or activity that is associated with a disease or disorder.
- affinity tag is meant any moiety used for the purification of a protein or nucleic acid molecule to which it is fixed.
- aggressive treatment regiment is meant a treatment regiment consistent with the treatment of a disease having an increased risk of lethality or a poor prognosis for recovery.
- treatments include chemotherapy, surgery, radiotherapy of increased dosage or toxicity relative to the treatment of a disease having a lower risk of lethality or a better prognosis.
- amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
- Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, for example, hydroxyproline, gamma-carboxyglutamate, and O-phosphoserine, phosphothreonine.
- amino acid analog a compound that has the same basic chemical structure as a naturally occurring amino acid, i.e., a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group (e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium), but that contains some alteration not found in a naturally occurring amino acid (e.g., a modified side chain);
- amino acid mimetic refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that function in a manner similar to a naturally occurring amino acid.
- Amino acid analogs may have modified R groups (for example, norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
- an amino acid analog is a D-amino acid, a ⁇ - amino acid, or an N-methyl amino acid.
- Amino acids and analogs are well known in the art. Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
- antibody is meant any immunoglobulin polypeptide, or fragment thereof, having immunogen binding ability.
- detectable amino acid sequence or “detectable moiety” is meant a composition that when linked with the nucleic acid or protein molecule of interest renders the latter detectable, via any means, including spectroscopic, photochemical, biochemical, immunochemical, or chemical means.
- useful labels include radioactive isotopes, magnetic beads, metallic beads, colloidal particles, fluorescent dyes, electron-dense reagents, enzymes (for example, as commonly used in an ELISA), biotin, digoxigenin, or haptens.
- a "labeled nucleic acid or oligonucleotide probe” is one that is bound, either covalently, through a linker or a chemical bond, or noncovalently, through ionic bonds, van der Waals forces, electrostatic attractions, hydrophobic interactions, or hydrogen bonds, to a label such that the presence of the nucleic acid or probe may be detected by detecting the presence of the label bound to the nucleic acid or probe.
- An "expression vector” is a nucleic acid construct, generated recombinantly or synthetically, bearing a series of specified nucleic acid elements that enable transcription of a particular gene in a host cell. Typically, gene expression is placed under the control of certain regulatory elements, including constitutive or inducible promoters, tissue-preferred regulatory elements, and enhancers.
- an effective amount is meant the amount of a required to ameliorate the symptoms of a disease relative to an untreated patient.
- the effective amount of active compound(s) used to practice the present invention for therapeutic treatment of a neoplastic disease varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the attending physician or veterinarian will decide the appropriate amount and dosage regimen. Such amount is referred to as an "effective" amount.
- fragment is meant a portion (e.g., at least 10, 25, 50, 100, 125, 150, 200, 250,
- the portion retains at least 50%, 75%, or 80%, or more preferably 90%, 95%, or even 99% of the biological activity of the reference protein or nucleic acid described herein.
- a "host cell” is any prokaryotic or eukaryotic cell that contains either a cloning vector or an expression vector. This term also includes those prokaryotic or eukaryotic cells that have been genetically engineered to contain the cloned gene(s) in the chromosome or genome of the host cell.
- inhibitory nucleic acid is meant a double-stranded RNA, siRNA (short interfering RNA), shRNA (short hairpin RNA), or antisense RNA, or a portion thereof, or a mimetic thereof, that when administered to a mammalian cell results in a decrease (e.g., by 10%, 25%, 50%, 75%, or even 90-100%) in the expression of a target gene.
- a nucleic acid inhibitor comprises at least a portion of a target nucleic acid molecule, or an ortholog thereof, or comprises at least a portion of the complementary strand of a target nucleic acid molecule.
- isolated refers to material that is free to varying degrees from components which normally accompany it as found in its native state.
- levels of purity may be applied as needed according to this invention in the different methodologies set forth herein; the customary purity standards known in the art may be used if no standard is otherwise specified.
- isolated nucleic acid molecule is meant a nucleic acid (e.g., a DNA, RNA, or analog thereof) that is free of the genes which, in the naturally-occurring genome of the organism from which the nucleic acid molecule of the invention is derived, flank the gene.
- the term therefore includes, for example, a recombinant DNA that is incorporated into a vector; into an autonomously replicating plasmid or virus; or into the genomic DNA of a prokaryote or eukaryote; or that exists as a separate molecule (for example, a cDNA or a genomic or cDNA fragment produced by PCR or restriction endonuclease digestion) independent of other sequences.
- the term includes an RNA molecule which is transcribed from a DNA molecule, as well as a recombinant DNA which is part of a hybrid gene encoding additional polypeptide sequence.
- “Microarray” means a collection of nucleic acid molecules or polypeptides from one or more organisms arranged on a solid support (for example, a chip, plate, or bead).
- neoplasia any disease that is caused by or results in inappropriately high levels of cell division, inappropriately low levels of apoptosis, or both.
- cancer is an example of a neoplasia.
- cancers include, without limitation, leukemias (e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, acute myeloblasts leukemia, acute promyelocyte leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelocytic leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphoma (Hodgkin's disease, non-Hodgkin's disease), Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma,
- nucleic acid is meant an oligomer or polymer of ribonucleic acid or deoxyribonucleic acid, or analog thereof. This term includes oligomers consisting of naturally occurring bases, sugars, and intersugar (backbone) linkages as well as oligomers having non-naturally occurring portions which function similarly. Such modified or substituted oligonucleotides are often preferred over native forms because of properties such as, for example, enhanced stability in the presence of nucleases.
- nucleic acids envisioned for this invention may contain phosphorothioates, phosphotriesters, methyl phosphonates, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar linkages.
- oligonucleotides having morpholino backbone structures are also preferred.
- the phosphodiester backbone of the oligonucleotide may be replaced with a polyamide backbone, the bases being bound directly or indirectly to the aza nitrogen atoms of the polyamide backbone (P. E. Nielsen et al. Science 199: 254, 1997).
- oligonucleotides may contain alkyl and halogen-substituted sugar moieties comprising one of the following at the 2' position: OH, SH, SCH3, F 5 OCN, O(CH2) n NH2 or O(CH2) n CH3, where n is from 1 to about 10; Ci to Cio lower alkyl, substituted lower alkyl, alkaryl or aralkyl; Cl; Br; CN; CF 3 ; OCF 3 ; O ⁇ , S-, or N-alkyl; O-, S-, or N-alkenyl; SOCH3; SO2CH3; ONO2; NO2; N3; NH2; heterocycloalkyl; heterocycloalkaryl; aminoalkylamino; polyalkylamino; substituted silyl; an RNA cleaving group; a conjugate; a reporter group; an intercalator; a group for improving the pharmacokinetic properties of an oligonucleotide;
- Oligonucleotides may also have sugar mimetics such as cyclobutyls in place of the pentofuranosyl group.
- Other preferred embodiments may include at least one modified base form.
- modified bases include 2-(amino)adenine, 2-(methylamino)adenine, 2- (imidazolylalkyl)adenine, 2-(aminoalklyamino)adenine, or other heterosubstituted alkyladenines.
- operably linked is meant that a first polynucleotide is positioned adjacent to a second polynucleotide that directs transcription of the first polynucleotide when appropriate molecules (e.g., transcriptional activator proteins) are bound to the second polynucleotide.
- ortholog is meant any protein or nucleic acid molecule of an organism that is highly related to a reference protein or nucleic acid sequence from another organism.
- recombinant is meant the product of genetic engineering or chemical synthesis.
- protein is meant any chain of amino acids, or analogs thereof, regardless of length or post-translational modification.
- positioned for expression is meant that the polynucleotide of the invention (e.g., a DNA molecule) is positioned adjacent to a DNA sequence that directs transcription and translation of the sequence (i.e., facilitates the production of, for example, a recombinant protein of the invention, or an RNA molecule).
- reference is meant a standard or control condition.
- siRNA is meant a double stranded RNA.
- an siRNA is 18, 19, 20, 21, 22, 23 or 24 nucleotides in length and has a 2 base overhang at its 3' end.
- These dsRNAs can be introduced to an individual cell or to a whole animal; for example, they may be introduced systemically via the bloodstream.
- Such siRNAs are used to downregulate mRNA levels or promoter activity.
- telomere binding molecule e.g., peptide, polynucleotide
- a sample for example, a biological sample, which naturally includes a protein of the invention.
- subject is meant a mammal, including, but not limited to, a human or non-human mammal, such as a bovine, equine, canine, ovine, or feline.
- substantially identical is meant a protein or nucleic acid molecule exhibiting at least 50% identity to a reference amino acid sequence (for example, any one of the amino acid sequences described herein) or nucleic acid sequence (for example, any one of the nucleic acid sequences described herein).
- a reference amino acid sequence for example, any one of the amino acid sequences described herein
- nucleic acid sequence for example, any one of the nucleic acid sequences described herein.
- such a sequence is at least 60%, more preferably 80% or 85%, and most preferably 90%, 95% or even 99% identical at the amino acid level or nucleic acid to the sequence used for comparison.
- Sequence identity is typically measured using sequence analysis software (for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs). Such software matches identical or similar sequences by assigning degrees of homology to various substitutions, deletions, and/or other modifications. Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. In an exemplary approach to determining the degree of identity, a BLAST program may be used, with a probability score between e'3 and e ⁇ 100 indicating a closely related sequence.
- sequence analysis software for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Bio
- transformed cell is meant a cell into which (or into an ancestor of which) has been introduced, by means of recombinant DNA techniques, a polynucleotide molecule encoding (as used herein) a protein of the invention.
- the invention features compositions and methods useful for the diagnosis of a neoplasia in a subject. These methods and compositions are based, in part, on the discovery that ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3) expression is increased in neoplastic tissues. In addition, the invention also provides methods and compositions for altering ASPM expression in a neoplastic cell. Such compositions and methods are likely to be useful for the treatment of neoplasia. Diagnostics
- Neoplastic tissues express higher levels of an ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3) or an ASPM polynucleotide than corresponding normal tissues. Accordingly, expression levels of an ASPM nucleic acid molecule or polypeptide are correlated with a particular disease state (e.g., neoplasia), and thus are useful in diagnosis. Accordingly, the present invention provides a number of diagnostic assays that are useful for the identification or characterization of a neoplasia.
- a patient having a neoplasia will show an increase in the expression of an ASPM nucleic acid molecule.
- Alterations in gene expression are detected using methods known to the skilled artisan and described herein. Such information can be used to diagnose a neoplasia.
- an alteration in the expression of an ASPM nucleic acid molecule is detected using real-time quantitative PCR (Q-rt-PCR) to detect changes in gene expression.
- Primers used for amplification of an ASPM nucleic acid molecule are useful in diagnostic methods of the invention.
- the primers of the invention embrace oligonucleotides of sufficient length and appropriate sequence so as to provide specific initiation of polymerization on a significant number of nucleic acids.
- the term "primer” as used herein refers to a sequence comprising two or more deoxyribonucleotides or ribonucleotides, preferably more than three, and most preferably more than 8, which sequence is capable of initiating synthesis of a primer extension product, which is substantially complementary to a locus strand.
- the primer must be sufficiently long to prime the synthesis of extension products in the presence of the inducing agent for polymerization.
- the exact length of primer will depend on many factors, including temperature, buffer, and nucleotide composition.
- the oligonucleotide primer typically contains between 12 and 27 or more nucleotides, although it may contain fewer nucleotides.
- Primers of the invention are designed to be "substantially" complementary to each strand of the genomic locus to be amplified and include the appropriate G or C nucleotides as discussed above. This means that the primers must be sufficiently complementary to hybridize with their respective strands under conditions that allow the agent for polymerization to perform.
- the primers should have sufficient complementarity with the 5' and 3' flanking sequences to hybridize therewith and permit amplification of the genomic locus. While exemplary primers are provided herein, it is understood that any primer that hybridizes with the target sequences of the invention are useful in the method of the invention for detecting ASPM nucleic acid molecules.
- ASPM-specific primers amplify a desired genomic target using the polymerase chain reaction (PCR). The amplified product is then detected using standard methods known in the art.
- a PCR product i.e., amplicon
- real-time PCR product is detected by probe binding.
- probe binding generates a fluorescent signal, for example, by coupling a fluorogenic dye molecule and a quencher moiety to the same or different oligonucleotide substrates (e.g., TaqMan® (Applied Biosystems, Foster City, CA, USA), Molecular Beacons (see, for example, Tyagi et al., Nature Biotechnology 14(3):303-8, 1996), Scorpions® (Molecular Probes Inc., Eugene, OR, USA)).
- a PCR product is detected by the binding of a fluorogenic dye that emits a fluorescent signal upon binding (e.g., SYBR® Green (Molecular Probes)). Such detection methods are useful for the detection of an ASPM PCR product.
- a fluorogenic dye molecule and a quencher moiety to the same or different oligonucleotide substrates (e.g., TaqMan® (Applied Biosystems, Foster City, CA, USA), Molecular Beacons (see, for example, Tyagi et al.
- hybridization with PCR probes that are capable of detecting an ASPM nucleic acid molecule, including genomic sequences, or closely related molecules, may be used to hybridize to a nucleic acid sequence derived from a patient having a neoplasia.
- the specificity of the probe determines whether the probe hybridizes to a naturally occurring sequence, allelic variants, or other related sequences.
- Hybridization techniques may be used to identify mutations indicative of a neoplasia, or may be used to monitor expression levels of these genes (for example, by Northern analysis (Ausubel et al., supra).
- humans may be diagnosed for a propensity to develop a neoplasia by direct analysis of the sequence of an ASPM nucleic acid molecule.
- the sequence of an ASPM nucleic acid molecule derived from a subject is compared to a reference sequence. An alteration in the sequence of the ASPM nucleic acid molecule relative to the reference indicates that the patient has or has a propensity to develop a neoplasia.
- diagnostic methods of the invention are used to assay the expression of an ASPM polypeptide in a biological sample relative to a reference (e.g., the level of ASPM polypeptide present in a corresponding control tissue).
- the level of an ASPM polypeptide is detected using an antibody that specifically binds an ASPM polypeptide.
- Exemplary antibodies that specifically bind an ASPM polypeptide are described herein. Such antibodies are useful for the diagnosis of a neoplasia.
- Methods for measuring an antibody- ASPM complex include, for example, detection of fluorescence, luminescence, chemiluminescence, absorbance, reflectance, transmittance, birefringence or refractive index.
- Optical methods include microscopy (both confocal and non-confocal), imaging methods and non-imaging methods. Methods for performing these assays are readily known in the art.
- Useful assays include, for example, an enzyme immune assay (EIA) such as enzyme-linked immunosorbent assay (ELISA), a radioimmune assay (RIA), a Western blot assay, or a slot blot assay. These methods are also described in, e.g., Methods in Cell Biology: Antibodies in Cell Biology, volume 37 (Asai, ed. 1993); Basic and Clinical Immunology (Stites & Terr, eds., 7th ed. 1991); and Harlow & Lane, supra. Immunoassays can be used to determine the quantity of ASPM in a sample, where an increase in the level of the ASPM polypeptide is diagnostic of a patient having a neoplasia.
- the measurement of an ASPM polypeptide or nucleic acid molecule in a subject sample is compared with a diagnostic amount present in a reference.
- a diagnostic amount distinguishes between a neoplastic tissue and a control tissue.
- the skilled artisan appreciates that the particular diagnostic amount used can be adjusted to increase sensitivity or specificity of the diagnostic assay depending on the preference of the diagnostician. In general, any significant increase (e.g., at least about 10%, 15%, 30%, 50%, 60%, 75%, 80%, or 90%) in the level of an ASPM polypeptide or nucleic acid molecule in the subject sample relative to a reference may be used to diagnose a neoplasia.
- the reference is the level of ASPM polypeptide or nucleic acid molecule present in a control sample obtained from a patient that does not have a neoplasia.
- the reference is a baseline level of ASPM present in a biologic sample derived from a patient prior to, during, or after treatment for a neoplasia.
- the reference is a standardized curve.
- the level of an ASPM polypeptide or nucleic acid molecule can be measured in different types of biologic samples.
- the biologic sample is a tissue sample that includes cells of a tissue or organ. Such tissue is obtained, for example, from a biopsy.
- the biologic sample is a biologic fluid sample (e.g., blood, blood plasma, serum, urine, seminal fluids, ascites, or cerebrospinal fluid).
- kits for the diagnosis or monitoring of a neoplasia in a biological sample obtained from a subject detects an increase in the expression of an ASPM nucleic acid molecule or polypeptide relative to a reference level of expression.
- the kit detects an alteration in the sequence of an ASPM nucleic acid molecule derived from a subject relative to a reference sequence.
- the kit includes reagents for monitoring the expression of an ASPM nucleic acid molecule, such as primers or probes that hybridize to an ASPM nucleic acid molecule.
- the kit includes an antibody that binds to an ASPM polypeptide.
- the kit includes directions for monitoring ASPM nucleic acid molecule or polypeptide levels in a biological sample derived from a subject.
- the kit comprises a sterile container which contains the primer, probe, antibody, or other detection regents; such containers can be boxes, ampules, bottles, vials, tubes, bags, pouches, blister-packs, or other suitable container form known in the art.
- Such containers can be made of plastic, glass, laminated paper, metal foil, or other materials suitable for holding nucleic acids.
- the instructions will generally include information about the use of the primers or probes described herein and their use in diagnosing a neoplasia.
- the kit further comprises any one or more of the reagents described in the diagnostic assays described herein.
- the instructions include at least one of the following: description of the primer or probe; methods for using the enclosed materials for the diagnosis of a neoplasia; precautions; warnings; indications; clinical or research studies; and/or references.
- the instructions may be printed directly on the container (when present), or as a label applied to the container, or as a separate sheet, pamphlet, card, or folder supplied in or with the container.
- the disease state or treatment of a patient having a neoplasia can be monitored using the methods and compositions of the invention.
- a microarray is used to assay the expression profile of an ASPM nucleic acid molecule. Such monitoring may be useful, for example, in assessing the efficacy of a particular drug in a patient.
- Therapeutics that alter the expression of an ASPM nucleic acid molecule or ASPM polypeptide are taken as particularly useful in the invention.
- Antibodies are well known to those of ordinary skill in the science of immunology.
- the term “antibody” means not only intact antibody molecules, but also fragments of antibody molecules that retain immunogen binding ability. Such fragments are also well known in the art and are regularly employed both in vitro and in vivo. Accordingly, as used herein, the term “antibody” means not only intact immunoglobulin molecules but also the well-known active fragments F(ab') 2 , and Fab. F(ab') 2 , and Fab fragments which lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding of an intact antibody (Wahl et al., J. Nucl. Med. 24:316-325 (1983).
- the antibodies of the invention comprise whole native antibodies, bispecific antibodies; chimeric antibodies; Fab, Fab', single chain V region fragments (scFv) and fusion polypeptides.
- an antibody that binds an ASPM polypeptide is monoclonal.
- the anti-ASPM antibody is a polyclonal antibody.
- the preparation and use of polyclonal antibodies are also known the skilled artisan.
- the invention also encompasses hybrid antibodies, in which one pair of heavy and light chains is obtained from a first antibody, while the other pair of heavy and light chains is obtained from a different second antibody. Such hybrids may also be formed using humanized heavy and light chains. Such antibodies are often referred to as "chimeric" antibodies.
- intact antibodies are said to contain "Fc” and "Fab” regions.
- the Fc regions are involved in complement activation and are not involved in antigen binding.
- An antibody from which the Fc' region has been enzymatically cleaved, or which has been produced without the Fc' region, designated an "F(ab') 2 " fragment retains both of the antigen binding sites of the intact antibody.
- an antibody from which the Fc region has been enzymatically cleaved, or which has been produced without the Fc region designated an "Fab"' fragment, retains one of the antigen binding sites of the intact antibody.
- Fab' fragments consist of a covalently bound antibody light chain and a portion of the antibody heavy chain, denoted "Fd.”
- the Fd fragments are the major determinants of antibody specificity (a single Fd fragment may be associated with up to ten different light chains without altering antibody specificity). Isolated Fd fragments retain the ability to specifically bind to immunogenic epitopes.
- Antibodies can be made by any of the methods known in the art utilizing ASPM polypeptides (e.g., ASPM, ASPM variant 1, 2, or 3), or immunogenic fragments thereof, as an immunogen.
- ASPM polypeptides e.g., ASPM, ASPM variant 1, 2, or 3
- immunogenic fragments thereof can be provided to the host in a delivery vehicle that is taken up by immune cells of the host.
- the cells will in turn express the receptor on the cell surface generating an immunogenic response in the host.
- nucleic acid sequences encoding an ASPM polypeptide e.g., ASPM, ASPM variant 1, 2, or 3
- immunogenic fragments thereof can be expressed in cells in vitro, followed by isolation of the receptor and administration of the receptor to a suitable host in which antibodies are raised.
- Antibody purification methods may include salt precipitation (for example, with ammonium sulfate), ion exchange chromatography (for example, on a cationic or anionic exchange column preferably run at neutral pH and eluted with step gradients of increasing ionic strength), gel filtration chromatography (including gel filtration HPLC), and chromatography on affinity resins such as protein A, protein G, hydroxyapatite, and antiimmunoglobulin.
- salt precipitation for example, with ammonium sulfate
- ion exchange chromatography for example, on a cationic or anionic exchange column preferably run at neutral pH and eluted with step gradients of increasing ionic strength
- gel filtration chromatography including gel filtration HPLC
- affinity resins such as protein A, protein G, hydroxyapatite, and antiimmunoglobulin.
- Antibodies can be conveniently produced from hybridoma cells engineered to express the antibody. Methods ofmaking hybridomas are well known in the art.
- the hybridoma cells can be cultured in a suitable medium, and spent medium can be used as an antibody source. Polynucleotides encoding the antibody of interest can in turn be obtained from the hybridoma that produces the antibody, and then the antibody may be produced synthetically or recombinantly from these DNA sequences. For the production of large amounts of antibody, it is generally more convenient to obtain an ascites fluid.
- the method of raising ascites generally comprises injecting hybridoma cells into an immunologically naive histocompatible or immunotolerant mammal, especially a mouse.
- the mammal may be primed for ascites production by prior administration of a suitable composition; e.g., Pristane.
- Monoclonal antibodies (Mabs) produced by methods of the invention can be "humanized” by methods known in the art.
- “Humanized” antibodies are antibodies in which at least part of the sequence has been altered from its initial form to render it more like human immunoglobulins. Techniques to humanize antibodies are particularly useful when non- human animal (e.g., murine) antibodies are generated. Examples of methods for humanizing a murine antibody are provided in U.S. Patent Nos. 4,816,567, 5,530,101, 5,225,539, 5,585,089, 5,693,762 and 5,859,205. ASPM Polypeptide Expression
- ASPM polypeptides, variants, and fragments thereof may be produced by transformation of a suitable host cell with all or part of a polypeptide-encoding nucleic acid molecule or fragment thereof in a suitable expression vehicle.
- suitable host cell e.g., E.
- coli or in a eukaryotic host (e.g., Saccharomyces cerevisiae, insect cells, e.g., S£21 cells, or mammalian cells, e.g., NIH 3T3, HeLa, or preferably COS cells).
- a eukaryotic host e.g., Saccharomyces cerevisiae, insect cells, e.g., S£21 cells, or mammalian cells, e.g., NIH 3T3, HeLa, or preferably COS cells.
- Such cells are available from a wide range of sources (e.g., the American Type Culture Collection, Rockland, Md.; also, see, e.g., Ausubel et al., supra).
- the method of transformation or transfection and the choice of expression vehicle will depend on the host system selected. Transformation and transfection methods are described, e.g., in Ausubel et al. (supra); expression vehicles may be chosen from those
- Expression vectors useful for producing such polypeptides include, without limitation, chromosomal, episomal, and virus-derived vectors, e.g., vectors derived from bacterial plasmids, from bacteriophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, and vectors derived from combinations thereof.
- virus-derived vectors e.g., vectors derived from bacterial plasmids, from bacteriophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retrovirus
- polypeptide production is the E. coli pET expression system (Novagen, Inc., Madison, Wis).
- E. coli pET expression system DNA encoding a polypeptide is inserted into a pET vector in an orientation designed to allow expression. Since the gene encoding such a polypeptide is under the control of the T7 regulatory signals, expression of the polypeptide is achieved by inducing the expression of T7 RNA polymerase in the host cell. This is typically achieved using host strains that express T7 RNA polymerase in response to IPTG induction.
- recombinant polypeptide is then isolated according to standard methods known in the art, for example, those described herein.
- pGEX expression system Another bacterial expression system for polypeptide production is the pGEX expression system (Pharmacia).
- This system employs a GST gene fusion system that is designed for high-level expression of genes or gene fragments as fusion proteins with rapid purification and recovery of functional gene products.
- the protein of interest is fused to the carboxyl terminus of the glutathione S-transferase protein from Schistosoma japonicum and is readily purified from bacterial lysates by affinity chromatography using Glutathione Sepharose 4B. Fusion proteins can be recovered under mild conditions by elution with glutathione.
- Cleavage of the glutathione S-transferase domain from the fusion protein is facilitated by the presence of recognition sites for site-specific proteases upstream of this domain.
- proteins expressed in pGEX-2T plasmids may be cleaved with thrombin; those expressed in pGEX-3X may be cleaved with factor Xa.
- the recombinant polypeptide of the invention is expressed, it is isolated, e.g., using affinity chromatography.
- an antibody e.g., produced as described herein
- a polypeptide of the invention may be attached to a column and used to isolate the recombinant polypeptide. Lysis and fractionation of polypeptide-harboring cells prior to affinity chromatography may be performed by standard methods (see, e.g., Ausubel et al., supra).
- the recombinant protein can, if desired, be further purified, e.g., by high performance liquid chromatography (see, e.g., Fisher, Laboratory Techniques In Biochemistry and Molecular Biology, eds., Work and Burdon, Elsevier, 1980).
- Polypeptides of the invention particularly short peptide fragments, can also be produced by chemical synthesis (e.g., by the methods described in Solid Phase Peptide Synthesis, 2nd ed., 1984 The Pierce Chemical Co., Rockford, 111.). These general techniques of polypeptide expression and purification can also be used to produce and isolate useful peptide fragments or analogs (described herein).
- ASPM Polypeptides and Analogs Also included in the invention are ASPM polypeptides, variants, or fragments thereof containing at least one alteration relative to a reference sequence. Such alterations include certain mutations, deletions, insertions, or post-translational modifications.
- the invention further includes analogs of any naturally-occurring polypeptide of the invention. Analogs can differ from naturally-occurring polypeptides of the invention by amino acid sequence differences, by post-translational modifications, or by both. Analogs of the invention will generally exhibit at least 85%, more preferably 90%, and most preferably 95% or even 99% identity with all or part of a naturally-occurring amino acid sequence of the invention.
- the length of sequence comparison is at least 10, 13, 15 amino acid residues, preferably at least 25 amino acid residues, and more preferably more than 35 amino acid residues.
- a BLAST program may be used, with a probability score between e "3 and e "100 indicating a closely related sequence.
- Modifications include in vivo and in vitro chemical derivatization of polypeptides, e.g., acetylation, carboxylation, phosphorylation, or glycosylation; such modifications may occur during polypeptide synthesis or processing or following treatment with isolated modifying enzymes. Analogs can also differ from the naturally-occurring polypeptides of the invention by alterations in primary sequence.
- the invention also includes fragments of any one of the polypeptides of the invention.
- a fragment means at least 5, 10, 13, or 15 amino acids.
- a fragment is at least 20 contiguous amino acids, at least 30 contiguous amino acids, or at least 50 contiguous amino acids, and in other embodiments at least 60 to 80 or more contiguous amino acids. Fragments of the invention can be generated by methods known to those skilled in the art or may result from normal protein processing (e.g., removal of amino acids from the nascent polypeptide that are not required for biological activity or removal of amino acids by alternative mRNA splicing or alternative protein processing events).
- ASPM Polynucleotides in general, includes any nucleic acid sequence encoding an ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3). Also included in the methods of the invention are any nucleic acid molecule containing at least one strand that hybridizes with such a nucleic acid sequence (e.g., an inhibitory nucleic acid molecule, such as a dsRNA, siRNA, shRNA, or antisense molecule). An isolated nucleic acid molecule can be manipulated using recombinant DNA techniques well known in the art.
- nucleotide sequence contained in a vector in which 5' and 3' restriction sites are known, or for which polymerase chain reaction (PCR) primer sequences have been disclosed is considered isolated, but a nucleic acid sequence existing in its native state in its natural host is not.
- An isolated nucleic acid may be substantially purified, but need not be.
- a nucleic acid molecule that is isolated within a cloning or expression vector may comprise only a tiny percentage of the material in the cell in which it resides. Such a nucleic acid is isolated, however, as the term is used herein, because it can be manipulated using standard techniques known to those of ordinary skill in the art.
- Polynucleotide therapy featuring a polynucleotide encoding an ASPM protein, variant, or fragment thereof is another therapeutic approach for treating a neoplasia.
- Such nucleic acid molecules can be delivered to cells of a subject having a neoplasia.
- the nucleic acid molecules must be delivered to the cells of a subject in a form in which they can be taken up so that therapeutically effective levels of an ASPM protein (e.g., ASPM, ASPM variant 1, 2, or 3) or fragment thereof can be produced.
- Transducing viral e.g., retroviral, adenoviral, and adeno-associated viral
- Transducing viral can be used for somatic cell gene therapy, especially because of their high efficiency of infection and stable integration and expression (see, e.g., Cayouette et al., Human Gene Therapy 8:423-430, 1997; Kido et al., Current Eye Research 15:833-844, 1996; Bloomer et al., Journal of Virology 71 :6641-6649, 1997; Naldini et al., Science 272:263-267, 1996; and Miyoshi et al., Proc. Natl. Acad. Sci. U.S.A. 94:10319, 1997).
- a polynucleotide encoding an ASPM protein, variant, or a fragment thereof can be cloned into a retroviral vector and expression can be driven from its endogenous promoter, from the retroviral long terminal repeat, or from a promoter specific for a target cell type of interest.
- viral vectors that can be used include, for example, a vaccinia virus, a bovine papilloma virus, or a herpes virus, such as Epstein-Barr Virus (also see, for example, the vectors of Miller, Human Gene Therapy 15-14, 1990; Friedman, Science 244:1275-1281, 1989; Eglitis et al., BioTechniques 6:608-614, 1988; Tolstoshev et al., Current Opinion in Biotechnology 1:55- 61, 1990; Sharp, The Lancet 337:1277-1278, 1991; Cornetta et al., Nucleic Acid Research and Molecular Biology 36:311-322, 1987; Anderson, Science 226:401-409, 1984; Moen, Blood Cells 17:407-416, 1991; Miller et al., Biotechnology 7:980-990, 1989; Le Gal La Salle et al., Science 259:988-990, 1993; and Johnson, Chest 107:77S-83S, 1995
- Retroviral vectors are particularly well developed and have been used in clinical settings (Rosenberg et al., N. Engl. J. Med 323:370, 1990; Anderson et al., U.S. Pat. No. 5,399,346). Most preferably, a viral vector is used to administer an ASPM polynucleotide systemically.
- Non- viral approaches can also be employed for the introduction of therapeutic to a cell of a patient diagnosed as having a neoplasia.
- a nucleic acid molecule can be introduced into a cell by administering the nucleic acid in the presence of lipofection (Feigner et al., Proc. Natl. Acad. Sci. U.S.A. 84:7413, 1987; Ono et al., Neuroscience Letters 17:259, 1990; Brigham et al., Am. J. Med. Sci.
- nucleic acids are administered in combination with a liposome and protamine.
- Gene transfer can also be achieved using non-viral means involving transfection in vitro. Such methods include the use of calcium phosphate, DEAE dextran, electroporation, and protoplast fusion. Liposomes can also be potentially beneficial for delivery of DNA into a cell. Transplantation of normal genes into the affected tissues of a patient can also be accomplished by transferring a normal nucleic acid into a cultivatable cell type ex vivo (e.g., an autologous or heterologous primary cell or progeny thereof), after which the cell (or its descendants) are injected into a targeted tissue.
- a cultivatable cell type ex vivo e.g., an autologous or heterologous primary cell or progeny thereof
- cDNA expression for use in polynucleotide therapy methods can be directed from any suitable promoter (e.g., the human cytomegalovirus (CMV), simian virus 40 (SV40), or metallothionein promoters), and regulated by any appropriate mammalian regulatory element.
- CMV human cytomegalovirus
- SV40 simian virus 40
- metallothionein promoters e.g., the human cytomegalovirus (CMV), simian virus 40 (SV40), or metallothionein promoters
- enhancers known to preferentially direct gene expression in specific cell types can be used to direct the expression of a nucleic acid.
- the enhancers used can include, without limitation, those that are characterized as tissue- or cell-specific enhancers.
- regulation can be mediated by the cognate regulatory sequences or, if desired, by regulatory sequences derived from a heterologous source, including any of the promoters or regulatory elements described above.
- a recombinant therapeutic such as a recombinant ASPM protein, variant, or fragment thereof
- ASPM protein recombinant ASPM protein
- variant, or fragment thereof a recombinant therapeutic that results in a recombinant recombinant recombinant recombinant recombinant recombinant recombinant recombinant recombinant recombinant recombinant ASPM protein, variant, or fragment thereof
- the dosage of the administered protein depends on a number of factors, including the size and health of the individual patient. For any particular subject, the specific dosage regimes should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions. Screening Assays
- an ASPM polypeptide As reported herein, the expression of an ASPM polypeptide is increased in neoplastic tissues. Accordingly, compounds that modulate the expression or activity of an ASPM polypeptide, variant, or fragment thereof are useful in the methods of the invention for the treatment or prevention of a neoplasm (e.g., breast, colon, lymph, ovary, stomach, thyroid, testis, and uterine cancer). Any number of methods are available for carrying out screening assays to identify such compounds. In one approach, candidate compounds are identified that specifically bind to and alter the activity of a polypeptide of the invention (e.g., an ASPM activity associated with cell proliferation, mitosis or maintenance of chromosomal stability). Methods of assaying such biological activities are known in the art and are described herein.
- a candidate compound may be tested in vitro for interaction and binding with a polypeptide of the invention and its ability to modulate cell proliferation, mitosis, or maintenance of chromosomal stability.
- ASPM's function in chromosomal stability can be assayed by detecting, for example, chromosomal nondysjunction in cells where endogenous ASPM expression is perturbed or reduced.
- Standard methods for perturbing or reducing ASPM expression include mutating or deleting an endogenous ASPM sequence, interfering with ASPM expression using RNAi, or microinjecting an ASPM-expressing cell with an antibody that binds ASPM and interferes with its function.
- chromosomal nondysjunction can be assayed in vivo, for example, in a mouse model in which ASPM has been knocked out by homologous recombination, or any other standard method.
- Potential agonists and antagonists of an ASPM polypeptide include organic molecules, peptides, peptide mimetics, polypeptides, nucleic acid molecules (e.g., double- stranded RNAs, siRNAs, antisense polynucleotides), and antibodies that bind to a nucleic acid sequence or polypeptide of the invention and thereby inhibit or extinguish its activity.
- Potential antagonists also include small molecules that bind to the ASPM polypeptide thereby preventing binding to cellular molecules with which the ASPM polypeptide normally interacts, such that the normal biological activity of the ASPM polypeptide is reduced or inhibited.
- Small molecules of the invention preferably have a molecular weight below 2,000 daltons, more preferably between 300 and 1,000 daltons, and most preferably between 400 and 700 daltons. It is preferred that these small molecules are organic molecules.
- a candidate compound that binds to an ASPM polypeptide, variant, or fragment thereof may be identified using a chromatography-based technique.
- a recombinant polypeptide of the invention may be purified by standard techniques from cells engineered to express the polypeptide (e.g., those described above) and may be immobilized on a column.
- a solution of candidate compounds is then passed through the column, and a compound specific for the ASPM polypeptide is identified on the basis of its ability to bind to the ASPM polypeptide and be immobilized on the column.
- the column is washed to remove non-specifically bound molecules, and the compound of interest is then released from the column and collected. Similar methods may be used to isolate a compound bound to a polypeptide microarray. Compounds isolated by this method (or any other appropriate method) may, if desired, be further purified (e.g., by high performance liquid chromatography).
- these candidate compounds may be tested for their ability to alter the biological activity of an ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3).
- any in vivo protein interaction detection system for example, any two-hybrid assay may be utilized to identify compounds that interact with an ASPM polypeptide. Interacting compounds isolated by this method (or any other appropriate , method) may, if desired, be further purified (e.g., by high performance liquid chromatography). Compounds isolated by any approach described herein may be used as therapeutics to treat a neoplasia in a human patient.
- compounds that inhibit the expression of an ASPM nucleic acid molecule whose expression is increased in a patient having a neoplasia are also useful in the methods of the invention. Any number of methods are available for carrying out screening assays to identify new candidate compounds that alter the expression of an ASPM nucleic acid molecule.
- candidate compounds are added at varying concentrations to the culture medium of cultured cells expressing one of the nucleic acid sequences of the invention. Gene expression is then measured, for example, by microarray analysis, Northern blot analysis (Ausubel et al., supra), or RT-PCR, using any appropriate fragment prepared from the nucleic acid molecule as a hybridization probe.
- the level of gene expression in the presence of the candidate compound is compared to the level measured in a control culture medium lacking the candidate molecule.
- a compound that promotes an alteration in the expression of an ASPM gene, or a functional equivalent thereof, is considered useful in the invention; such a molecule may be used, for example, as a therapeutic to treat a neoplasia in a human patient.
- the effect of candidate compounds is measured at the level of polypeptide production to identify those that promote an alteration in an ASPM polypeptide level.
- the level of ASPM polypeptide can be assayed using any standard method. Standard immunological techniques include Western blotting or immunoprecipitation with an antibody specific for an ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3).
- ASPM an antibody specific for an ASPM polypeptide
- ASPM antigen specific for an ASPM polypeptide
- ASPM ASPM, ASPM variant 1, 2, or 3
- immunoassays may be used to detect or monitor the expression of at least one of the polypeptides of the invention in an organism.
- Polyclonal or monoclonal antibodies produced as described above may be used in any standard immunoassay format (e.g., ELISA, Western blot, or RIA assay) to measure the level of the polypeptide.
- a compound that promotes a decrease in the expression or biological activity of the polypeptide is considered particularly useful. Again, such a molecule may be used, for example, as a therapeutic to delay, ameliorate, or treat a neoplasia in a human patient.
- a nucleic acid described herein is expressed as a transcriptional or translational fusion with a detectable reporter, and expressed in an isolated cell (e.g., mammalian or insect cell) under the control of a heterologous promoter, such as an inducible promoter.
- the cell expressing the fusion protein is then contacted with a candidate compound, and the expression of the detectable reporter in that cell is compared to the expression of the detectable reporter in an untreated control cell.
- a candidate compound that alters the expression of the detectable reporter is a compound that is useful for the treatment of a neoplasia. In one embodiment, the compound decreases the expression of the reporter.
- Each of the DNA sequences listed herein may also be used in the discovery and development of a therapeutic compound for the treatment of neoplasia.
- the encoded protein upon expression, can be used as a target for the screening of drugs.
- the DNA sequences encoding the amino terminal regions of the encoded protein or Shine-Delgarno or other translation facilitating sequences of the respective mRNA can be used to construct sequences that promote the expression of the coding sequence of interest. Such sequences may be isolated by standard techniques (Ausubel et al., supra).
- the invention also includes novel compounds identified by the above-described screening assays.
- such compounds are characterized in one or more appropriate animal models to determine the efficacy of the compound for the treatment of a neoplasia.
- characterization in an animal model can also be used to determine the toxicity, side effects, or mechanism of action of treatment with such a compound.
- novel compounds identified in any of the above-described screening assays may be used for the treatment of a neoplasia in a subject. Such compounds are useful alone or in combination with other conventional therapies known in the art.
- compounds capable of inhibiting the growth or proliferation of a neoplasia by altering the expression or biological activity of an ASPM polypeptide, variant, or fragment thereof are identified from large libraries of either natural product or synthetic (or semisynthetic) extracts or chemical libraries according to methods known in the art. Numerous methods are also available for generating random or directed synthesis (e.g., semi-synthesis or total synthesis) of any number of chemical compounds, including, but not limited to, saccharide-, lipid-, peptide-, and nucleic acid-based compounds. Synthetic compound libraries are commercially available from Brandon Associates (Merrimack, N.H.) and Aldrich Chemical (Milwaukee, Wis.).
- test compounds of the invention are present in any combinatorial library known in the art, including: biological libraries; peptoid libraries (libraries of molecules having the functionalities of peptides, but with a novel, non-peptide backbone which are resistant to enzymatic degradation but which nevertheless remain bioactive; see, e.g., Zuckermann, R.N. et al, J.
- compounds shown to be useful agents for the treatment of a neoplasm are chemically modified according to methods known in the art.
- Therapeutics useful in the methods of the invention include, but are not limited to, those that have an anti-neoplastic activity or those that alter an ASPM biological activity associated with cell proliferation or maintenance of chromosomal stability.
- Neoplastic cell growth is not subject to the same regulatory mechanisms that govern the growth or proliferation of normal cells.
- Compounds that reduce the growth or proliferation of a neoplasm are useful for the treatment of neoplasms. Methods of assaying cell growth and proliferation are known in the art. See, for example, Kittler et al. (Nature. 432 (7020): 1036- 40, 2004) and by Miyamoto et al. (Nature 416(6883):865-9, 2002).
- Assays for cell proliferation generally involve the measurement of DNA synthesis during cell replication.
- DNA synthesis is detected using labeled DNA precursors, such as ([ 3 H]- Thymidine or 5-bromo-2'-deoxyuridine [BrdU], which are added to cells (or animals) and then the incorporation of these precursors into genomic DNA during the S phase of the cell cycle (replication) is detected (Ruefli-Brasse et al., Science 302(5650):1581-4, 2003; Gu et al., Science 302 (5644):445-9, 2003).
- labeled DNA precursors such as ([ 3 H]- Thymidine or 5-bromo-2'-deoxyuridine [BrdU]
- Candidate compounds that reduce the survival of a neoplastic cell are also useful as anti-neoplasm therapeutics.
- Assays for measuring cell viability are known in the art, and are described, for example, by Crouch et al. (J. Immunol. Meth. 160, 81-8); Kangas et al. (Med. Biol.62, 338-43, 1984); Lundin et al., (Meth. Enzymol.133, 27-42, 1986); Petty et al.
- Cell viability can be assayed using a variety of methods, including MTT (3- (4,5-dimethylthiazolyl)-2,5-diphenyltetrazolium bromide) (Barltrop, Bioorg. & Med. Chem. Lett.1: 611, 1991; Cory et al., Cancer Comm. 3, 207-12, 1991; Paull J. Heterocyclic Chem. 25, 911, 1988). Assays for cell viability are also available commercially.
- CELLTITER-GLO ® Luminescent Cell Viability Assay Promega
- CellTiter-Glo ® Luminescent Cell Viability Assay which is a lactate dehyrodgenase (LDH) cytotoxicity assay.
- LDH lactate dehyrodgenase
- Candidate compounds that increase neoplastic cell death are also useful as anti-neoplasm therapeutics. Assays for measuring cell apoptosis are known to the skilled artisan.
- Apoptotic cells are characterized by characteristic morphological changes, including chromatin condensation, cell shrinkage and membrane blebbing, which can be clearly observed using light microscopy.
- the biochemical features of apoptosis include DNA fragmentation, protein cleavage at specific locations, increased mitochondrial membrane permeability, and the appearance of phosphatidylserine on the cell membrane surface.
- Assays for apoptosis are known in the art. Exemplary assays include TUNEL (Terminal deoxynucleotidyl Transferase Biotin-dUTP Nick End Labeling) assays, caspase activity (specifically caspase-3) assays, and assays for fas-ligand and annexin V.
- the methods of the invention may also be used for microarray-based assays that provide for the high-throughput analysis of cancer biomarkers.
- the ASPM nucleic acid molecules or polypeptides of the invention are useful as hybridizable array elements in such a microarray.
- the array elements are organized in an ordered fashion such that each element is present at a specified location on the substrate.
- Useful substrate materials include membranes, composed of paper, nylon or other materials, filters, chips, glass slides, and other solid supports. The ordered arrangement of the array elements allows hybridization patterns and intensities to be interpreted as expression levels of particular genes or proteins.
- nucleic acid microarray oligonucleotides may be synthesized or bound to the surface of a substrate using a chemical coupling procedure and an ink jet application apparatus, as described in PCT application WO95/251116 (Baldeschweiler et al.), incorporated herein by reference.
- a gridded array may be used to arrange and link cDNA fragments or oligonucleotides to the surface of a substrate using a vacuum system, thermal, UV, mechanical or chemical bonding procedure.
- a nucleic acid molecule derived from a biological sample may be used to produce a hybridization probe as described herein.
- the biological samples are generally derived from a patient, preferably as a bodily fluid (such as blood, cerebrospinal fluid, phlegm, saliva, or urine) or tissue sample (e.g. a tissue sample obtained by biopsy). For some applications, cultured cells or other tissue preparations may be used.
- tissue sample e.g. a tissue sample obtained by biopsy.
- cultured cells or other tissue preparations may be used.
- the mRNA is isolated according to standard methods, and cDNA is produced and used as a template to make complementary RNA suitable for hybridization. Such methods are described herein.
- RNA is amplified in the presence of fluorescent nucleotides, and the labeled probes are then incubated with the microarray to allow the probe sequence to hybridize to complementary oligonucleotides (e.g., ASPM nucleic acid molecules) bound to the microarray.
- complementary oligonucleotides e.g., ASPM nucleic acid molecules
- stringent salt concentration will ordinarily be less than about 750 mM NaCl and 75 mM trisodium citrate, preferably less than about 500 mM NaCl and 50 mM trisodium citrate, and most preferably less than about 250 mM NaCl and 25 mM trisodium citrate.
- Low stringency hybridization can be obtained in the absence of organic solvent, e.g., formamide, while high stringency hybridization can be obtained in the presence of at least about 35% formamide, and most preferably at least about 50% formamide.
- Stringent temperature conditions will ordinarily include temperatures of at least about 30 0 C, more preferably of at least about 37°C, and most preferably of at least about 42°C. Varying additional parameters, such as hybridization time, the concentration of detergent, e.g., sodium dodecyl sulfate (SDS), and the inclusion or exclusion of carrier DNA, are well known to those skilled in the art. Various levels of stringency are accomplished by combining these various conditions as needed. In one embodiment, hybridization will occur at 3O 0 C in 750 mM NaCl, 75 mM trisodium citrate, and 1% SDS.
- SDS sodium dodecyl sulfate
- hybridization will occur at 37°C in 500 mM NaCl, 50 mM trisodium citrate, 1% SDS, 35% formamide, and 100 ⁇ g/ml denatured salmon sperm DNA (ssDNA). In yet another embodiment, hybridization will occur at 42°C in 250 mM NaCl, 25 mM trisodium citrate, 1% SDS, 50% formamide, and 200 ⁇ g/ml ssDNA. Useful variations on these conditions will be readily apparent to those skilled in the art.
- wash stringency conditions can be defined by salt concentration and by temperature. As above, wash stringency can be increased by decreasing salt concentration or by increasing temperature.
- stringent salt concentration for the wash steps will preferably be less than about 30 mM NaCl and 3 mM trisodium citrate, and most preferably less than about 15 mM NaCl and 1.5 mM trisodium citrate.
- Stringent temperature conditions for the wash • steps will ordinarily include a temperature of at least about 25°C, at least about 42°C, or at least about 68 0 C.
- wash steps will occur at 25°C in 30 mM NaCl, 3 mM trisodium citrate, and 0.1% SDS. In another embodiment, wash steps will occur at 42°C in 15 mM NaCl, 1.5 mM trisodium citrate, and 0.1% SDS. In yet another embodiment, wash steps will occur at 68°C in 15 mMNaCl, 1.5 mM trisodium citrate, and 0.1% SDS. Additional variations on these conditions will be readily apparent to those skilled in the art.
- a detection system may be used to measure the absence, presence, and amount of hybridization for all of the distinct sequences simultaneously (e.g., Heller et al., Proc. Natl. Acad. Sci. 94:2150-2155, 1997).
- a scanner is used to determine the levels and patterns of fluorescence.
- ASPM polypeptides may also be analyzed using protein microarrays. Such arrays are useful in high- throughput low-cost screens to identify peptide or candidate compounds that bind a polypeptide of the invention, or fragment thereof.
- protein microarrays feature a protein, or fragment thereof, bound to a solid support.
- Suitable solid supports include membranes (e.g., membranes composed of nitrocellulose, paper, or other material), polymer- based films (e.g., polystyrene), beads, or glass slides.
- ASPM polypeptides e.g., ASPM, ASPM variant 1, 2, or 3
- ASPM polypeptides are spotted on a substrate using any convenient method known to the skilled artisan (e.g., by hand or by inkjet printer).
- ASPM antibody binding e.g., ASPM antibody binding
- the protein microarray is hybridized with a detectable probe.
- probes can be polypeptide (e.g., an ASPM antibody), nucleic acid, or small molecules.
- polypeptide and nucleic acid probes are derived from a biological sample taken from a patient, such as a bodily fluid (such as blood, urine, saliva, or phlegm); a homogenized tissue sample (e.g. a tissue sample obtained by biopsy); or cultured cells (e.g., lymphocytes).
- Probes can also include antibodies, candidate peptides, nucleic acids, or small molecule compounds derived from a peptide, nucleic acid, or chemical library.
- Hybridization conditions e.g., temperature, pH, protein concentration, and ionic strength
- Such conditions are known to the skilled artisan and are described, for example, in Harlow, E. and Lane, D., Using Antibodies: A Laboratory Manual. 1998, New York: Cold Spring Harbor Laboratories.
- specifically bound probes are detected, for example, by fluorescence, enzyme activity (e.g., an enzyme-linked calorimetric assay), direct immunoassay, radiometric assay, or any other suitable detectable method known to the skilled artisan.
- ASPM detection may be combined with the detection of other biomarkers, where the presence or level of the biomarker is correlated with the presence of a neoplasia.
- the present invention contemplates pharmaceutical preparations comprising an ASPM protein, a polynucleotide that encodes an ASPM protein, or an ASPM inhibitory nucleic acid molecule (e.g., a polynucleotide that hybridizes to and interferes with the expression of an ASPM polynucleotide), together with a pharmaceutically acceptable carrier.
- Polynucleotides of the invention may be administered as part of a pharmaceutical composition.
- the compositions should be sterile and contain a therapeutically effective amount of the polypeptides or nucleic acid molecules in a unit of weight or volume suitable for administration to a subject.
- compositions ordinarily will be stored in unit or multi-dose containers, for example, sealed ampoules or vials, as an aqueous solution or as a lyophilized formulation for reconstitution.
- a lyophilized formulation 10 mL vials are filled with 5 mL of sterile-filtered 1% (w/v) aqueous ASPM polynucleotide solution, such as an aqueous solution of ASPM polynucleotide or polypeptide, and the resulting mixture can then be lyophilized.
- the infusion solution can be prepared by reconstituting the lyophilized material using sterile Water-for-Injection (WFI).
- WFI Water-for-Injection
- the ASPM polynucleotide, or polypeptide, or analogs may be combined, optionally, with a pharmaceutically acceptable excipient.
- pharmaceutically-acceptable excipient means one or more compatible solid or liquid filler, diluents or encapsulating substances that are suitable for administration into a human.
- carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate administration.
- the components of the pharmaceutical compositions also are capable of being co-mingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction that would substantially impair the desired pharmaceutical efficacy.
- compositions can be administered in effective amounts.
- the effective amount will depend upon the mode of administration, the particular condition being treated and the desired outcome. It may also depend upon the stage of the condition, the age and physical condition of the subject, the nature of concurrent therapy, if any, and like factors well known to the medical practitioner. For therapeutic applications, it is that amount sufficient to achieve a medically desirable result.
- an effective amount is sufficient to stabilize, slow, or reduce the proliferation of the neoplasm.
- doses of active polynucleotide compositions of the present invention would be from about 0.01 mg/kg per day to about 1000 mg/kg per day. It is expected that doses ranging from about 50 to about 2000 mg/kg will be suitable. Lower doses will result from certain forms of administration, such as intravenous administration. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits. Multiple doses per day are contemplated to achieve appropriate systemic levels of the ASPM polynucleotide or polypeptide compositions of the present invention.
- a variety of administration routes are available.
- the methods of the invention may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects.
- Other modes of administration include oral, rectal, topical, intraocular, buccal, intravaginal, intracisternal, intracerebroventricular, intratracheal, nasal, transdermal, within/on implants, e.g., fibers such as collagen, osmotic pumps, or grafts comprising appropriately transformed cells, etc., or parenteral routes.
- a particular method of administration involves coating, embedding or derivatizing fibers, such as collagen fibers, protein polymers, etc. with therapeutic proteins.
- Other useful approaches are described in Otto, D. et al., J. Neurosci. Res. 22: 83-91 and in Otto, D. and Unsicker, K. J. Neurosci. 10: 1912-1921.
- compositions and methods of the invention may be used in combination with any conventional therapy known in the art.
- an ASPM polynucleotide or polypeptide composition of the invention having anti-neoplastic activity may be used in combination with any anti-neoplastic therapy known in the art.
- Exemplary anti-neoplastic therapies include, for example, chemotherapy, cryotherapy, hormone therapy, radiotherapy, and surgery.
- a ASPM polynucleotide composition of the invention may, if desired, include one or more chemotherapeutics typically used in the treatment of a neoplasm, such as abiraterone acetate, altretamine, anhydrovinblastine, auristatin, bexarotene, bicalutamide, BMSl 84476, 2,3,4,5,6-pentafluoro-N-(3-fluoro-4-methoxyphenyl)benzene sulfonamide, bleomycin, N,N-dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-proly- l-Lproline-t- butylamide, cachectin, cemadotin, chlorambucil, cyclophosphamide, 3',4'-didehydro-4'- deoxy- ⁇ '-norvin- caleukoblastine, docetaxol, doxetax
- Primary autosomal recessive microcephaly is a genetic disorder in which an affected individual has a head circumference >3 standard deviations below the age- and sex-related mean and is mentally retarded, but typically has no other abnormal findings. Brain scans show that while the entire brain is reduced in size, the cerebral cortex is most severely affected. Because the vast majority of neurons are generated by week 21 of fetal life, microcephaly is likely due to a reduced production of neurons. The most common cause of microcephaly appears to be mutations in the ASPM gene.
- the ASPM gene which spans 65 kb of genomic DNA at Iq31, encodes a 10,434 base pair coding sequence (GenBank Accession No. NM_018136) containing 28 exons.
- the predicted ASPM protein has two distinguished regions, a tandem pair of N- terminal putative calponin-homology (CH) domains and a large block of IQ motifs (the single letter codes for the amino acids isoleucine (I) and glutamine (Q)), a sequence which mediates interactions with calmodulin and calmodulin-related proteins.
- CH domains are about 100 residues long and are commonly involved in actin binding, but many other substrates are found in vivo (Gimona et al., FEBS Lett. 513, 98-106, 2002; Korenbaum et al., J. Cell Sci. 115, 3543-3545, 2002).
- the IQ calmodulin-binding motif comprises 20-25 amino acids, with the core fitting the consensus IQXXXRGXXXR (where X is any amino acid) (Mooseker et al., Annual. Rev. Cell. Dev. Biol. 11, 633-675, 1995; Houdusse et al., Structure 4, 1475-1490, 1996).
- CH and IQ domains were first discovered in motor proteins such as unconventional myosins (Matin et al., Protein Sci. 12, 2909-2923).
- Calmodulin binding to IQ-motifs induces a conformational change in the proteins that regulates the binding of actin to the amino- terminal CH domains (Bahler et al., FEBS Lett.
- myosins typically contain between 2 and 6 IQ motifs. They are not identical within a single protein, i.e. some of them have a higher affinity for the Ca +2 -free form of calmodulin, while other motifs have a higher affinity for the Ca +2 /calmodulin.
- the number of IQ motifs and their divergence seems to determine the length of the lever arm and hence the step-size of myosin motor.
- ASPM does not have the catalytic Sl motor domain and therefore it cannot function as a mechanochemical protein.
- ASPM also differs from all known motor proteins by an extreme abundance of IQ motifs.
- the protein contains 81 putative calmodulin-binding motifs, most of which are encoded by exon 18, the largest exon in ASPM as reported herein.
- the predicted ASPM protein is conserved between mammals, Drosophila, and worm, with a consistent correlation between nervous system complexity and protein length, principally involving an increase in the number of encoded IQ domains.
- the Caenorhabditis elegans ASP homologue of ASPM contains 2 IQ domains; the Drosophila melonagaster ASP protein contains 24 repeats, and there are between 60-81 IQ domains in mammals.
- Drosophila abnormal spindle ( ⁇ spj-deficient mutants exhibit a mitotic metaphase checkpoint arrest with abnormal spindle poles, suggesting that Asp is required for the integrity of microtubule organizing centers.
- the Drosophila asp gene encodes a 220 kDa microtubule-associated protein found at the spindle poles and centrosomes from prophase to early telophase.
- the Asp protein has consensus phosphorylation sites for CDKl and MAP kinases (Saunders et al., J. Cell. Biol. 137, 881-890, 1997).
- Asp is co-purified with ⁇ — tubulin from centrosomes and they are both required for the organization of microtubules into asters (Avides et al., Science 283, 1733-1735, 1999). This activity is dependent on the phosphorylation of Asp by the kinase Polo (Avides et al., Nat. Cell. Biol. 4, 421-424, 2001; Glover et al., Oncogene 24, 230-237, 2005). Together these observations suggest that mutations in human ASPM may cause microcephaly due to the disregulation of mitotic spindle activity in neuronal progenitor cells.
- ASPM and alternatively spliced isoforms of ASPM are expressed in a variety of embryonic and adult tissues and are upregulated in neoplasia.
- Example 1 ASPM is expressed in embryonic and adult tissues
- ASPM1F/ASPM28R S'-tcctgtctctcagccacttc-S'/S 1 - 10,315 bp or 5,560 bp* atgccaagcgtatccatcac-3'
- Drosophila asp homologue is expressed in some adult tissues
- human ASPM expression was analyzed in a variety of adult tissues, including breast, lung, pancreas, uterus, colon, thyroid, liver, bladder, kidney, ovary, testis, stomach, lymph node cervix, esophagus, and brain.
- Human ASPM transcripts were detected in all tissues except adult brain, though the level of expression was much lower than was observed in fetal tissues.
- the ASPM expression analysis suggested that ASPM functions in non-CNS (non-central nervous system) tissues.
- Example 2 ASPM expression is upregulated in cancer Expression data was obtained from over 120 uterine cancers and gene expression was analyzed using high density oligonucleotide microarrays. This analysis identified ASPM as one of several genes highly overexpressed in uterine cancer as compared to normal endometrium. Based on this initial observation, ASPM expression in normal and cancer tissues was analyzed by quantitative real-time RT-PCR. These studies confirmed that ASPM is more highly expressed in cancer of the uterus and ovary as compared to their expression in corresponding normal tissues ( Figures 2A and 2B).
- ASPM expression was checked by RT-PCR using primers FN2-3 and RN2-3 developed from exon 2 and exon 3 sequences (see Table 1).
- the ASPM gene is widely expressed in a variety of adult and embryo tissues and is upregulated in malignancy. Without being tied to one particular theory, it is possible that ASPM is upregulated in malignancy, because its expression correlates with the number of dividing cells in the malignant tissue.
- ASPM may be required in tumors that are increasing their size just as it is required for increases in brain size in microcephalic individuals.
- Example 3 Alternatively spliced variants of human ASPM encode different numbers of IQ domains
- the open reading frame (ORF) of ASPM was predicted based on the sequences of multiple small size Expressed Sequence Tags (ESTs) deposited into GenBank, which cover 28 exons.
- ESTs Expressed Sequence Tags
- RT-PCR analysis was carried out using primers, ASPMlF and ASPM28R, which are present in the presumed first and last exons of ASPM (Table 1).
- Figured 4A and 4B actin
- two predominant PCR products were detected in RNA from fetal tissues. The largest of these RNAs migrated at a size consistent with the predicted full-length mRNA of 10,434 base pairs and was present in all fetal human tissues analyzed. The next most abundant band migrated at approximately 5.6 kb.
- FIG. 3A Expression of mouse ASPM in normal murine tissues is shown in Figure 3B.
- the predominant PCR products were directly sequenced.
- the largest human PCR product corresponded to the predicted full-length ORF of 10,434 nucleotides.
- Further cloning and sequencing of the smaller RTPCR fragments revealed three human mRNAs with open reading frames (ORFs) of 5,678 bp, 4,259 bp and 3,189 bp (GenBank Accession numbers AY971956, AY971957 and AY971955) (Table 3, and Figures 8C-8H). Wild-type ASPM polypeptide and polynucleotide sequences are shown in Figures 8 A and 8B.
- the ASPM gene may encode at least three isoforms ( Figure 5A and 5B) containing 1892 (variant 1 corresponding to the abundant RT-PCR product), 1389 (variant 2) and 1062 (variant 3) amino acid residues ( Figures 8C-8H).
- Figures 8A and 8B The sequence of the wild-type human ASPM polypeptide and the ASPM polynucleotide is provided at Figures 8A and 8B, respectively.
- the amino acid and nucleic acid sequence of the ASPM variants is provided at Figures 8C-8H.
- variant 1 lacks exon 18 (carrying 61 IQ domains) leaving this isoform with 14 total IQ domains.
- variant 2 and 3 utilize different splice junctions within exon 18 and truncate parts of the IQ array. As a result of this splicing, variant 2 has 41 complete plus one incomplete IQ domains and variant 3 has 27 complete and one incomplete IQ domain.
- both variants 2 and 3 lack exons 4-17, which encode two CH (calponin-homology) domains and variant 2 also lacks exon 27, and has a stop codon in the intronic region after exon 26.
- Example 4 IQ motifs in the ASPM protein are organized into a higher order repeat structure
- More than half of the human ASPM protein consists of repeated calmodulin-binding IQ domains.
- the IQ array which spans amino acid positions 1273-3234, is formed by 81 distinct IQ motifs of variable length ( Figures 6A-6C).
- Figures 6A-6C the IQ array present in mouse and rat exhibits one large deletion corresponding to a region between IQs 57-70 in human, and several smaller insertions and deletions.
- 67 IQ repeats are found in mouse Aspm ( Figure 6D).
- the numbers of IQ repeats are slightly higher than previously reported, 74 and 61 repeats for human and mouse, respectively (Bond et al., Nat. Genet. 32, 316-320, 2005; Bond et al., Am. J. Hum.
- the long IQ 6 repeat is followed by four short units, IQ 11 and IQ 15 by three units.
- the next 12 long units are in each case followed by two short units.
- This region forms a highly regular array of 63 aa long superunits, composed of one long 27 aa unit and two short 23 aa units.
- the N-terminal and C-terminal parts of the IQ repeat region are less regular and contain IQ units of variable lengths.
- the mouse Aspm protein displays the same periodicity 27-23-23 in the central region of the protein.
- the N and C terminals of the protein are less organized as is the case of human ASPM ( Figure 6D).
- the IQ consensus sequence includes two positively charged amino acids, the IQ-containing region that spans 2,000 amino acids is highly positively charged. Among 530 charged residues, 467 are arginine and lysine. This density of basic residues may be required for binding of calmodulin, and/or it may facilitate interactions between ASPM and acidic proteins.
- Example 5 Identification of a novel ASNP repeat region within ASPM
- ASPM ASPM N-terminal part of the protein.
- the repeats are 32 and 35 amino acids long and localized to positions 316-347 and 366-400, respectively ( Figures 5A and 5B). These repeats are highly conserved near the termini, with the central part being less well conserved. Similar repeats were found in all mammalian ASPM homologues. A single repeat was identified in the chicken gene. The interspecies conservation is similar to intraspecies comparisons, i.e. the termini are highly similar, the N- terminus is almost identical, but the central part is variable. The identified repeats are referred to as ASNP (for ASPM N-proximal) repeats. No significant similarity was identified between this motif and other proteins, even when using very sensitive searches (Position-
- Example 6 ASPM is found at the spindle poles during mitosis in human culture cells The role of Drosophila Asp in nucleating microtubules at centrosomes is consistent with its localization at the spindle poles during mitotic periods (Saunders et al., J. Cell. Biol. 137, 881-890, 1997).
- Polyclonal antibodies against three epitopes of human ASPM were generated.
- the anti-ASPM antibodies for two different epitopes, VTRK and QSPE, were used for indirect immunostaining of human HT1080 cultured cells.
- the VTRK antibody showed strong and similar typical spindle pole staining pattern during prometaphase, metaphase and anaphase ( Figures 7A-I).
- VTRK antibody A characterization of the VTRK antibody is shown at Figure 7 J, 7K, and 7L. A similar staining pattern was observed with the QSPE antibody during mitotic periods. The results of the mitotic subcellular localization and the highly conserved structural homology between Drosophila Asp and human ASPM suggested that human ASPM may be involved in a conserved spindle function.
- human ASPM is a mitotic spindle protein that is likely a functional orthologue of the Drosophila asp protein Bond et al., Nat. Genet. 32, 316-320 (6). This conclusion is supported by a recent analysis of proteins of the purified human mitotic spindles Sauer et al., (2005) Proteome analysis of the human mitotic spindle. MoI. Cell. Prot ⁇ omics 4, 35-43 (23) which provides evidence that microcephaly arises from deficient neurogenic mitosis.
- ASPM controls the proliferative symmetry of progenitors required for the expansion of cerebral cortical size Chenn et al., Cereb. Cortex 13, 599-606 (24).
- ASPM controls the proliferative symmetry of progenitors required for the expansion of cerebral cortical size Chenn et al., Cereb. Cortex 13, 599-606 (24).
- the presence of a nonfunctional ASPM might reduce chromosome segregation fidelity, resulting in a high incidence of chromosome aneuploidy, and reducing the ability of fetal stem cells to produce neurons.
- the Drosophila gene abnormal spindle encodes a novel microtubule- associated protein that associates with the polar regions of the mitotic spindle. J. Cell. Biol. 137, 881-890 (18, 19, 21).
- ASPM encodes several alternatively spliced mRNAs.
- the main ASPM isoform is a 3477 amino acid residue protein that contains eighty-one IQ motifs. Most of these are organized into a Higher-Order trimer Repeat (HOR) containing two units each comprising 23 and 27 amino acid residues.
- HOR Higher-Order trimer Repeat
- myosin V is an unconventional myosin that transports cellular cargos such as vesicles, melanosomes, or mRNA on actin filaments.
- the IQ periodicity seems to be necessary for efficient interaction between myosin V IQ domains and actin half-repeats, which are 36-nm long Sakamoto et al., J. Boil. Chem. 278, 29201-29207 (26).
- the ASPM IQ periodicity may provide the exact spacing required for interactions with polymeric periodical proteins such as actin.
- the major alternatively spliced form contains an in frame deletion of the entire exon 18 sequence and encodes 1892 amino acid residues with the predicted protein harboring fourteen flanking IQ domains that do not exhibit any periodicity. Similarly, two predominant transcripts were also present in mouse. In addition, alternatively spliced variants lacking both CH domains and more than a half of mostly periodical IQ domains were also detected, suggesting that ASPM may encode several proteins with different functions.
- the Drosophila gene abnormal spindle encodes a novel microtubule-associated protein that associates with the polar regions of the mitotic spindle.
- a specific role of IQ repeats may be accumulation of Ca +2 /calmodulins at the central region of the protein. Potentially one molecule of ASPM protein can bind through calmodulin several hundred calcium ions. Release of the bind Ca +2 may signal microtubule polymerization.
- Non-radioactive in situ hybridization was performed using a digoxigenin (DIG)- labelled cRNA probe.
- the antisense probe was generated from a mouse EST clone (Genbank accession: AW558815) using standard methods, and frozen sections hybridized and visualized using methods described previously (Berger et al., Jour. Comp. Neurol. 433, 101- 114, 2001).
- RNAs from fetal (brain, bladder, colon, testis, liver, heart, liver, skeletal muscle, skin, spleen, stomach), adult (brain, breast, lung, pancreas, uterus, colon, thyroid, liver, bladder, kidney, ovary, testis, stomach, lymph node, cervix, esophagus) and matching cancer human tissues, mouse (brain, testis, liver, heart) tissues (Ambion) were used for screening ASPM expression with the primers described in Table 1.
- cDNA was made from 1 ⁇ g of total RNA using a commercially available kit for reverse transcriptase polymerase chain reaction (RT-PCR), SUPERSCRIPT l s ⁇ STRAND SYSTEM kit (Invitrogen, Carlsbad, CA), and priming with oligo dT per their standard protocol.
- Human ⁇ -actin primers (BD Biosciences Clontech, Palo Alto, CA) were used as positive controls for both human and mouse RT-PCR.
- NCI-60 cancer cell lines were from the National Cancer Institute, NIH.
- RT-PCR was performed using 1 ⁇ l of cDNA in a 50 ul reaction volume.
- Standard reaction conditions were: 94°C 5m, (94°C 1 minute, 55°C 1 minute, 72°C 1 minute x 35 cycles), 72°C 7 minutes, 4°C hold. Sequencing of the RT-PCR products confirmed their identity to the ASPM transcripts.
- ASPM was assayed by quantitative PCR using a commercially available assay system, FAM- labeled TaqMan® Gene Expression Assays, from Applied Biosystems, (Foster City, CA). Detectably labeled ⁇ -actin, VIC-labeled ⁇ -actin (4326315E), was used as a reference. Samples were run on the ABI Prism® 7700 Sequence Detection System according to manufacturer's suggested Protocols in separate tubes. The relative quantitation, using the comparative Cx method, was calculated for each sample.
- the full-length ASPM transcripts (10,434 bp for human and 9,846 bp for mouse) were sequenced after cloning of RT-PCR products into a TA cloning vector (Invitrogen, Carlsbad, CA).
- the RT-PCR products corresponding to human ASPM alternatively spliced variants of 5,679 bp, 4,259 bp and 3,189 bp and mouse spliced variant of 5,574 bp were also sequenced. Sequence forward and reverse reactions were run on a PE- Applied Biosystem 3100
- the human ASPM protein was initially screened for IQ repeats by Radar fhttp://www.ebi.ac.uk/Radar/).
- the detected IQ repeats were used to build a hidden Markov profile using publically available software, HMMER v 2.2 Eddy, Bioinformatics 14, 755-763 (34); http://hmmer.wustl.edu/.
- the obtained profile was used to search for the remaining IQ repeats.
- the IQ sequences were manually edited in order to obtain exact boundaries of all IQ units. This complete set was used to build a new hidden Markov profile, which subsequently served for detection of IQ repeats in mouse Aspm.
- Other ASPM repeats, including newly detected N-terminal repeats were identified by Radar.
- Peptide specific antibodies and Western blot analysis were identified by Radar.
- the concentrates were then subjected to gel filtration chromatography using 2.6 x 60 cm SUPERDEX 200 columns (GE Healthcare, Piscataway, NJ) and the monomeric IgG fractions were pooled and concentrated.
- the protein concentrations were then determined using a commercially available kit, the Bradford assay (Bio-Rad, Richmond, CA).
- These peptides were identical to both human and mouse ASPM proteins.
- the antibodies specificity for these peptides was first confirmed with recombinant ASPM fragments. Plasmids for ASPM expression in E.coli cells were constructed by insertion of a 1353 bp fragment containing QSPE and VTKR epitopes, and a 705 bp fragment containing SRLK epitope.
- the fragments were PCR amplified from the full-size ASPM cDNA (positions 346-1695 and 9730-10434) and cloned into Bat ⁇ BI site of the pMAL-p2X expression vector (BioLabs Inc.).
- human (HTl 080) and mouse (3T3) cells were mixed with SDS sample buffer containing a protease inhibitor cocktail (Sigma), homogenized using a 27 gauge needle and resolved in a 3.4% of 29:1 acrylamide/bis-acrylamide gel.
- the proteins were transferred to PVDF membranes (Millipore) for 40 minutes at 15V in transfer buffer (50 mM Tris, 380 mM glycine, 0.1% SDS and 20% methanol) using a semi-dry method. All subsequent steps were carried out in phosphate buffered saline (PBS) containing 0.05% Tweet' 20 (TPBS). After thirty minutes blocking with 10% TPBS nonfat milk, the membranes were exposed for 12 hours to 1/2,000 diluted anti-QSPE or SRLK antibodies.
- transfer buffer 50 mM Tris, 380 mM glycine, 0.1% SDS and 20% methanol
- PBS phosphate buffered saline
- TPBS Tweet' 20
- the PVDF membrane was washed three times with TPBS, then incubated with anti-rabbit IgG conjugated with horse radish peroxidase (HRP) diluted 1/5000 for 30 minutes, and then washed as in the previous step.
- the membranes were incubated for 1 minute with commercially available chemiluminescent detection reagents, ECL plus reagents (Amersham) and exposed to Hperfilm ECL (Amersham).
- Immunoblotting of cellular proteins from human HTl 080 and mouse 3T3 cells revealed the predicted protein isoforms corresponding to two main alternatively spliced variants.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Molecular Biology (AREA)
- Pathology (AREA)
- Analytical Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Zoology (AREA)
- Urology & Nephrology (AREA)
- Oncology (AREA)
- Biotechnology (AREA)
- Microbiology (AREA)
- Physics & Mathematics (AREA)
- Hospice & Palliative Care (AREA)
- Wood Science & Technology (AREA)
- Genetics & Genomics (AREA)
- Biochemistry (AREA)
- Hematology (AREA)
- Biomedical Technology (AREA)
- General Health & Medical Sciences (AREA)
- General Engineering & Computer Science (AREA)
- Cell Biology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biophysics (AREA)
- Food Science & Technology (AREA)
- Medicinal Chemistry (AREA)
- General Physics & Mathematics (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
- Peptides Or Proteins (AREA)
Abstract
The invention features diagnostic and therapeutic methods and compositions featuring ASPM proteins and nucleic acid molecules whose expression is increased in neoplastic tissues.
Description
MITOTIC SPINDLE PROTEIN ASPM AS A DIAGNOSTIC MARKER FOR NEOPLASIA AND USES THEREFOR
CROSS-REFERENCE TO RELATED APPLICATION This application claims the benefit of the following U.S. Provisional Application No.:
60/696,212, which was filed on July 1, 2005, the contents of which are incorporated herein by reference.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
Research supporting this application was carried out by the United States of America as represented by the Secretary, Department of Health and Human Services.
BACKGROUND OF THE INVENTION Nearly 23 % of deaths in the United States in 2001 were cancer related (American
Cancer Society, 2001 Statistics). Gynecological cancers, such as uterine carcinoma and ovarian cancer are critical problems for women. Uterine cancer is the most common cancer in the United States among women. Ovarian cancer ranks fifth in cancer deaths among women, totaling more deaths than any other cancer of the female reproductive system. This may reflect the lack of a diagnostic test capable of diagnosing ovarian cancer in its early stages when it is most amenable to treatment. The effective treatment of ovarian cancer will likely be significantly increased if a screening test to identify early stage ovarian cancer is developed. Given the high incidence of ovarian cancer and other neoplasias, there is an urgent need for new diagnostic and therapeutic methods.
SUMMARY OF THE INVENTION
In general, the present invention provides for diagnostic methods, compositions, and kits that are useful for identifying a neoplasia by measuring Abnormal SPindle-like Microcephaly associated (ASPM) expression in a patient sample. In addition, the invention provides for methods of treating a neoplasia having increased ASPM expression.
In one aspect, the invention generally features a method of diagnosing a subject (e.g., a human) as having, or having a propensity to develop, a neoplasia (e.g., cancer of the brain, breast, lung, pancreas, uterus, colon, thyroid, liver, bladder, kidney, ovary, testis, stomach, lymph node, cervix, and esophagus). The method involves determining the level of expression of an ASPM nucleic acid molecule (e.g., any nucleic acid sequence provided
herein) in a subject sample, where an increased level of expression relative to a reference, indicates that the subject has or has a propensity to develop a neoplasia.
In another aspect, the invention features a method of diagnosing a subject as having, or having a propensity to develop, a neoplasia. The method involves determining the level of expression of an ASPM polypeptide (e.g., any amino acid sequence provided herein) in a subject sample, where an increased level (e.g., increased by at least 5%, 10%, 25%, 50%, 75%, 85%, 90% or 95%; or where the increase is by 5X, 10X, 2OX or 30X) of expression relative to the level of expression in a reference, indicates that the subject has or has a propensity to develop a neoplasia. In one embodiment, the method involves determining the level of expression of the ASPM polypeptide (e.g., using an immunological assay).
In another aspect, the invention features a method of diagnosing a subject as having, or having a propensity to develop, a neoplasia. The method involves determining the level of biological activity (e.g., mitotic spindle activity, calmodulin binding activity, cell proliferative active, maintenance of chromosomal stability) of an ASPM polypeptide in a subject sample, where an alteration in the level of biological activity relative to the biological activity in a reference, indicates that the subject has or has a propensity to develop a neoplasia.
In yet another aspect, the invention features a method of monitoring a subject diagnosed as having a neoplasia. The method involves determining the level of an ASPM polypeptide in a subject sample, where an alteration in the level of expression relative to the level of expression in a reference indicates the severity of neoplasia in the subject. In one embodiment, the subject is being treated for a neoplasia (e.g. breast, prostate, lung, testis, ovary, or uterine neoplasia). In one embodiment, the alteration is an increase or a decrease. In a related embodiment, the increase indicates an increased severity of neoplasia. In another embodiment, the reference is a control subject sample (e.g., a biological sample, such as a biological fluid or tissue sample). In one embodiment, the reference is a sample obtained at an earlier time point.
In various embodiments of the above aspects, the method is used to diagnose a subject as having neoplasia; is used to determine the treatment regimen for a subject having neoplasia; is used to monitor the condition of a subject being treated for neoplasia; or is used to determine the prognosis of a subject having neoplasia. In a related embodiment, a poor prognosis determines an aggressive treatment regimen for the subject.
In another aspect, the invention provides a method for identifying a subject as having or having a propensity to develop a neoplasia. The method involves detecting an alteration in
the sequence of an ASPM nucleic acid molecule relative to the sequence or expression of a reference molecule. In one embodiment, the alteration is detected using a hybridization reaction or is detected by sequencing the nucleic acid molecule.
In another aspect, the invention provides an ASPM antibody that specifically binds to an ASPM protein or fragment thereof (e.g., a VTRK. or QSPE epitope of an ASPM polypeptide).
In yet another aspect, the invention features a polypeptide containing an isolated ASPM protein variant, or fragment thereof, having substantial identity to ASPM variant 1, 2, or 3, where the polypeptide has an ASPM biological activity (e.g., mitotic spindle activity, calmodulin binding activity, functions in cell proliferation, or functions in chromosomal stability). In one embodiment, the ASPM protein variant is at least 80%, 85%, 90%, or 95% identical to ASPM variant 1, 2, or 3. In another embodiment, the ASPM protein variant contains at least an IQ domain and is capable of binding calmodulin. In yet another embodiment, the polypeptide consists of an ASPM protein variant selected from the group consisting of ASPM variant 1, 2, and 3. In various embodiments, the polypeptide is a fusion protein; is linked to a detectable amino acid sequence; or is linked to an affinity tag.
In another aspect, the invention features an isolated ASPM nucleic acid molecule, where the nucleic acid molecule encodes a polypeptide of any of the previous aspects, a vector comprising such a nucleic acid molecule, and a host cell (e.g., in vitro or in vivo) comprising the vector or nucleic acid molecule.
In a related aspect, the invention features an isolated ASPM inhibitory nucleic acid molecule, where the inhibitory nucleic acid molecule specifically binds at least a fragment of a nucleic acid molecule encoding an ASPM protein; a vector containing a nucleic acid molecule encoding the ASPM inhibitory nucleic acid molecule; and a host cell (e.g., in vitro or in vivo) comprising the vector or nucleic acid molecule.
In various embodiments of the previous aspects, the vector is an expression vector where the nucleic acid molecule is positioned for expression. In other embodiments, the nucleic acid molecule is operably linked to a promoter (e.g., a promoter suitable for expression in a mammalian cell). In yet other embodiments of any of the above aspects, the host cell expresses an ASPM protein variant. In still other embodiments, the host cell is a mammalian cell, such as a human cell.
In a related aspect, the invention features a double-stranded RNA corresponding to at least a portion of an ASPM nucleic acid molecule that encodes an ASPM protein, where the
double-stranded RNA is capable of altering protein expression level. In one embodiment, the
RNA is an siRNA.
In another related aspect, the invention features an antisense nucleic acid molecule, where the antisense nucleic acid molecule is complementary to at least six nucleotides of an ASPM nucleic acid molecule that encodes an ASPM protein, and where the antisense is capable of altering expression from the nucleic acid molecule to which it is complementary. In another aspect, the invention features a primer capable of binding to an ASPM nucleic acid molecule encoding an ASPM protein variant that is upregulated in a neoplastic tissue. In a related aspect, the invention features a collection of primers capable of binding to and amplifying an ASPM nucleic acid molecule, where at least one of the primers in the collection is the primer of the previous aspect. In one embodiment, the collection features at least one pair of primers capable of amplifying an ASPM protein (e.g., ASPM, ASPM variant
1, 2, or 3). In another embodiment, the primers are useful in diagnosing a neoplasia. In yet another aspect, the invention features a pharmaceutical composition containing an effective amount of an ASPM protein, variant, or fragment thereof, in a pharmaceutically acceptable excipient, where the fragment is capable of modulating (i.e., increasing or decreasing) cell proliferation or chromosome stability.
In yet another aspect, the invention features a pharmaceutical composition containing an effective amount of a nucleic acid molecule of any previous aspect in a pharmaceutically acceptable excipient, where the fragment is capable of modulating cell proliferation or chromosomal stability.
In yet another aspect, the invention features a pharmaceutical composition containing an effective amount of a vector containing a nucleic acid molecule encoding an ASPM protein of any previous aspect in a pharmaceutically acceptable excipient, where expression of the ASPM protein in a cell is capable of reducing, stabilizing, or inhibiting a neoplasia, decreasing cell proliferation or enhancing chromosomal stability. In one embodiment, the
ASPM nucleic acid molecule is positioned for expression in a mammalian cell.
In another aspect, the invention features an ASPM biomarker purified on a biochip. In yet another aspect, the invention features a microarray containing at least two nucleic acid molecules, or fragments thereof, fixed to a solid support, where at least one of the nucleic acid molecules is an ASPM nucleic acid molecule.
In a related aspect, the invention features a microarray containing at least two polypeptides, or fragments thereof, bound to a solid support, where at least one of the polypeptides on the support is an ASPM polypeptide.
In yet another aspect, the invention features a diagnostic kit for the diagnosis of a neoplasia in a subject containing an ASPM nucleic acid molecule, or fragment thereof, and written instructions for use of the kit for detection of a neoplasia.
In yet another aspect, the invention features a diagnostic kit for the diagnosis of a neoplasia in a subject containing an antibody that specifically binds an ASPM polypeptide, or fragment thereof, and written instructions for use of the kit for detection of a neoplasia. In yet another aspect, the invention features a kit identifying a subject as having or having a propensity to develop a neoplasia, containing an adsorbent, where the adsorbent retains an ASPM biomarker, and written instructions for use of the kit for detection of a neoplasia.
In yet another aspect, the invention features a kit containing a first capture reagent that specifically binds an ASPM biomarker, and written instructions for use of the kit for detection of a neoplasia.
In yet another aspect, the invention features a method for detecting neoplasia in a subject sample, where the method involves (a) contacting a subject sample with a capture reagent affixed to a substrate; and (b) capturing an ASPM polypeptide or nucleic acid molecule with the capture reagent. In one embodiment, the subject sample comprises an ASPM protein or fragment thereof. In another embodiment, the ASPM protein is fractionated prior to contacting the capture reagent.
In yet another aspect, the invention features a method of altering the expression of an ASPM nucleic acid molecule in a cell, the method comprising contacting the cell with an effective amount of a compound capable of altering the expression of the ASPM nucleic acid molecule. In one embodiment, the compound is an ASPM antisense nucleic acid molecule, a small interfering RNA (siRNA), or a double stranded RNA (dsRNA) that inhibits the expression of an ASPM nucleic acid molecule.
In yet another aspect, the invention features a method of altering ASPM protein expression in a cell, the method comprising contacting the cell with a compound capable of altering the expression of an ASPM polypeptide.
In various embodiments of the above aspects, the cell is a human cell, a neoplastic cell, a cell is in vitro or a cell in vivo.
In yet another aspect, the invention features a method of treating or preventing a neoplasia. The method involves administering to a subject (e.g., a human) in need thereof an effective amount of a pharmaceutical composition that alters expression of an ASPM polypeptide. In yet another aspect, the invention features a method of identifying a compound that inhibits a neoplasia. The method involves contacting a cell that expresses an ASPM nucleic acid molecule with a candidate compound, and comparing the level of expression of the nucleic acid molecule in the cell contacted by the candidate compound with the level of expression in a control cell not contacted by the candidate compound, where an alteration in expression of the ASPM nucleic acid molecule identifies the candidate compound as a compound that inhibits a neoplasia.
In a related aspect, the invention features a method of identifying a compound that inhibits a neoplasia. The method involves contacting a cell that expresses an ASPM polypeptide with a candidate compound, and comparing the level of expression of the polypeptide in the cell contacted by the candidate compound with the level of polypeptide expression in a control cell not contacted by the candidate compound, where an alteration in the expression of the ASPM polypeptide identifies the candidate compound as a compound that inhibits a neoplasia.
In another related aspect, the invention features a method of identifying a compound that inhibits a neoplasia. The method involves contacting a cell that expresses an ASPM polypeptide with a candidate compound, and comparing the biological activity of the polypeptide in the cell contacted by the candidate compound with the level of biological activity in a control cell not contacted by the candidate compound, where an alteration in the biological activity of the ASPM polypeptide identifies the candidate compound as a candidate compound that inhibits a neoplasia.
In another related aspect, the invention features method of identifying a candidate compound that inhibits a neoplasia, the method comprising a) contacting a cell containing an ASPM nucleic acid molecule present in an expression vector that includes a reporter construct; b) detecting the level of reporter gene expression in the cell contacted with the candidate compound with a control cell not contacted with the candidate compound, where an alteration in the level of the reporter gene expression identifies the candidate compound as a candidate compound that inhibits a neoplasia.
In various embodiments of the above aspects, the alteration in expression is an increase or a decrease in transcription. In other embodiments, the alteration in expression is
an increase or a decrease in translation. In still other embodiments, the cell is in vitro or in vivo. In still other embodiments, the cell is a human cell, such as a neoplastic cell. In yet other embodiments, the alteration in expression is assayed using an immunological assay, an enzymatic assay, or a radioimmunoassay. In various embodiments of the above aspects, the cell is a human cell, a neoplastic cell, a cell is in vitro or a cell in vivo. In various embodiments of any of the above-aspects, ASPM protein or nucleic acid molecule expression is increased in a neoplastic cell by at least 2X, 5X, 10X, 2OX, 30X, 4OX, 5OX the level of expression in a corresponding control cell. In various embodiments of any of the above aspects, a method or composition that alters (e.g., increases or decreases) ASPM expression by at least 5%, 10%, 25%, 50%, 75% or 100% is useful in the invention.
BRIEF DESCRIPTION OF THE DRAWINGS
Figures IA and IB show mouse and human ASPM expression in a variety of tissues. Figure IA is a photomicrograph showing ASPM expression in an E14 whole mouse embryo as detected by hybridization to antisense probe. Tissues are denoted using the following identifiers: Th=thalamus; Ma==mammilary region; H=heart; Li=liver; Lu=lung; KHkidney. Figure IB is a photograph of an agarose gel showing relative levels of human ASPM expression in embryonic tissues. The ASPM transcripts in various tissues were analyzed using RT-PCR. Expression analyses of the mRNA was performed using human multiple fetal tissue cDNAs. β-actin was used as an internal control.
Figures 2A and 2B are graphs showing expression of ASPM ia ovarian and uterine cancers. Figure 2A shows ASPM expression as determined using real-time PCR in samples of normal ovary (n=4) and ovarian cancers (n=48). Figure 2B shows ASPM expression as determined using real-time PCR in samples of normal endometrium (n=6), endometrioid endometrial cancer(n=15), mixed mesodermal tumors (MMT) of the uterus (n=15), and serous endometrial carcinomas (n=15).
Figures 3 A and 3B show RT-PCR expression of ASPM in various human and mouse tissues. Figure 3A shows expression in human normal and matching tumor tissues. Figure 4B shows expression in normal murine tissues. RT-PCR expression of actin is provided as a reference.
Figures 4A and 4B are photographs of an agarose gel containing alternatively spliced ASPM variants in fetal tissues (Figure 4A) and β-actin (Figure 4B) visualized using ethidium bromide. Two major ASPM transcripts with sizes of ~10.3 and 5.7 kb were identified in all
tissues analyzed. Additional spliced variants with variable sizes are also seen, β-actin was used as the internal control.
Figures 5A and 5B show ASPM spliced forms in human and mouse. Figure 5A is a schematic diagram that shows the positions of major domains in the ASPM protein. The putative microtubule-binding domain is in gray, the calponin-homology domain in orange (positions 960-1056 and 1114-1174), IQ repeats in blue, and the terminal domain in black. Yellow color marks positions of newly identified 32 and 35 as long repeats in the N-terminus (see B). The middle part shows major spliced variants and comparison with the full-length ASPM protein. The bottom part shows parts of the human ASPM protein encoded by individual exons. Spliced variant 2 contains exon 26 followed by a part of intron 26 where the ORF is disrupted by stop codon. To better separate individual exons, the odd numbered exons are colored in black and even numbered ones in white. Figure 5B shows sequence alignments among mammalian species for two ASPM repeats identified herein (SEQ ID Nos: 1-16). The human ASPM protein contains two repeats in positions 316-347 and 366-400. Thee repeats are found in other mammals and one is also preserved in chicken. Identical amino acids are identified in black boxes. Residues shown in upper case font are amino acids that are invariant among the species in the alignment. Residues shown in lower case font are amino acids that are conserved, but that are not invariant. Gray shading identifies conservative substitutions. Figures 6A-6D show the structure and evolution of human ASPM IQ repeats (SEQ ID
Nos: 17-97) (Figures 6 A, 6B, and 6C) and mouse IQ repeats Figure 6D. Figure 6 A shows the organization of IQ repeats in the human ASPM protein. At the far left of Figure 6A, the number of IQ repeats and their positions in the full-length ASPM protein is shown. The length of the individual repeat is also indicated by amino acid residue numbers. The human alternatively spliced variants are highlighted in orange. Note that variants 2 and 3 contain large deletions that extend further toward the C-terminus of ASPM. The blue box displays the region deleted in mouse and rat. Color is used to mark positions that are variable in the analyzed primates (green monkey, rhesus monkey, orangutan, gorilla, chimpanzee, and human) and also changes specific to African hominoids. Based on the Gonnet PAM250 matrix, substitutions were divided into noncoservative (P < 0.5), and conservative (the rest). The IQ repeats 4-54 form an organized array of longer (-27 aa) and shorter (~23 aa) units. Alignment and conservation is shown separately for long (Figure 6B) and short repeats (Figure 6C) from the IQ4-54 region. For both alignments, particular features (i.e., aliphatic,
polar, hydrophobic, positive, and small) of the most conserved amino acids positions are indicated in Figures 6B and 6C. Figure 6D shows the structure and evolution of mouse ASPM IQ repeats. The left column shows the number of IQ repeats and their positions in the full-length ASPM protein; the right column shows the length of the individual repeats. The grey positions mark sites that are different between mouse and rat proteins.
Figures 7A-7I are photomicrographs showing the cytological analysis of ASPM proteins in mitotic cells HT1080 cells. HT1080 cells grown on coverslips were analyzed by indirect immunofluorescent staining using antibodies against human ASPM (Figures 7B, 7E, and 7H). Chromosomal DNA was counterstained with DAPI (Figures 7A, 7D and 7G). Anti-ASPM staining shows characteristic spindle pole staining patterns during mitotic periods, prometaphase (VTRK), metaphase (QSPE) and anaphase (VTRK). Scale bar indicates 10 μm.
Figures 7 J, 7K, and 7L show a characterization of the anti-VTKR antibody. Figure 7A shows the analysis of protein extracts from a normal and a microcephalic individuals that were run on an SDS PAGE gel, transferred to a membrane, and immunoblotted with the anti- VTKR antibody. The Western blot in Figure 7 J shows the presence of two predicted ASPM proteins (a full-size 410 kDa protein and a 218 kDa isoform lacking an exon 18-encoded segment) in a protein extract derived from normal human cells (HT1080) (human lane). The predicted isoforms are 35 kDa shorter (i.e., 385 kDa and 183 kDa) in MM10458 cell protein extracts, which are derived from a microcephalic individual (patient lane). ASPM is truncated by a frameshift in exon 24 in MMl 0458. The anti-VTKR antibody also visualizes at least three additional bands in the HT 1080 extract; two of these are missing in the MMl 0458 extract. These bands may correspond to the predicted 164 kDa and 124 kDa ASPM isoforms. Figure 7K shows an immunoblot analysis of mouse ASPM proteins using the anti-VTKR antibody. The largest immunoreactive band in the mouse cell extract apparently corresponds to the predicted full-size 364 kDa ASPM protein that is 46 kDa shorter than human ASPM. The second major band apparently corresponds to the predicted 212 kDa mouse ASPM isoform lacking the exon 18-encoding segment that is approximately the same size as human ASPM isoform (218 kDa). Note that the small size bands visible in Figure 7J (i.e., < 150 kDa) were run off the gel to see the difference between the full-size human ASPM and mouse ASPM proteins. Figure 7L is a Western blot analysis showing the specificity of the affinity-purified anti-VTKR antibody. The antibody recognizes a recombinant MBP-ASPM fusion protein expressed in the pMAL-p2X expression vector.
Protein extract (100 μg) from bacterial cells was analyzed by SDS-PAGE, followed by immunoblotting using either anti-VTKR antibody (lanes 1-4) or anti-MBP antibody (lanes 5- 8). Lanes 1 and 5 correspond to the vector without insert. The predicted 60 kDa band corresponding to the fusion protein was detected. Lanes 2, 3, 4, 6, 7, and 8 correspond to individual transformants.
Figures 8A-8J shows the sequences of the human ASPM polypeptide (Figure 8A), its variants 1 (Figure 8C), 2 (Figure 8E), and 3 (Figure 8G), and the nucleic acid molecules encoding each of these polypeptides (Figures 8B, 8D, 8F, 8H). Figures 81 and 8 J provide the nucleic acid and amino acid sequences of murine Aspm.
DETAILED DESCRIPTION OF THE INVENTION Definitions
Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs. The following references provide one of skill with a general definition of many of the terms used in this invention: Singleton et al., Dictionary of Microbiology and Molecular Biology (2nd ed. 1994); The Cambridge Dictionary of Science and Technology (Walker ed., 1988); The Glossary of Genetics, 5th Ed., R. Rieger et al. (eds.), Springer Verlag (1991); and Hale & Marham, The Harper Collins Dictionary of Biology (1991). As used herein, the following terms have the meanings ascribed to them below, unless specified otherwise.
By "ASPM polypeptide" is meant a protein or protein variant, or fragment thereof, that is substantially identical to at least a portion of GenBank Accession No. NP__060606 (human ASPM) and that has an ASPM biological activity (e.g., calmodulin binding, function in mitosis, cell proliferation, regulation of symmetric cell division, or chromosomal stability). By "ASPM nucleic acid molecule" is meant a polynucleotide encoding an ASPM polypeptide or variant, or fragment thereof.
By "ASPM biological activity" is meant calmodulin binding activity, function during cell division, maintenance of chromosome stability, regulation of symmetric cell division, or any other function at a mitotic spindle, function in a neoplastic cell, or ASPM antibody binding.
By "alteration" is meant a change relative to a reference. For example, a change (increase or decrease) in the expression levels of a gene or polypeptide as detected by standard art known methods such as those described above. As used herein, an alteration
includes a 10% change in expression levels, preferably a 25% change, more preferably a 40% change, and most preferably a 50%, 75%, 85%, 95% or greater change in expression levels. "
By "biomarker" is meant any protein or polynucleotide having an alteration in expression level or activity that is associated with a disease or disorder. By "affinity tag" is meant any moiety used for the purification of a protein or nucleic acid molecule to which it is fixed.
By "aggressive treatment regiment" is meant a treatment regiment consistent with the treatment of a disease having an increased risk of lethality or a poor prognosis for recovery. Such treatments include chemotherapy, surgery, radiotherapy of increased dosage or toxicity relative to the treatment of a disease having a lower risk of lethality or a better prognosis.
The term "amino acid" refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, for example, hydroxyproline, gamma-carboxyglutamate, and O-phosphoserine, phosphothreonine.
By an "amino acid analog" is meant a compound that has the same basic chemical structure as a naturally occurring amino acid, i.e., a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group (e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium), but that contains some alteration not found in a naturally occurring amino acid (e.g., a modified side chain); the term " amino acid mimetic" refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that function in a manner similar to a naturally occurring amino acid. Amino acid analogs may have modified R groups (for example, norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. In one embodiment, an amino acid analog is a D-amino acid, a β- amino acid, or an N-methyl amino acid.
Amino acids and analogs are well known in the art. Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
By "antibody" is meant any immunoglobulin polypeptide, or fragment thereof, having immunogen binding ability.
In this disclosure, "comprises," "comprising," "containing" and "having" and the like can have the meaning ascribed to them in U.S. Patent law and can mean " includes,"
"including," and the like; "consisting essentially of or "consists essentially" likewise has the meaning ascribed in U.S. Patent law and the term is open-ended, allowing for the presence of more than that which is recited so long as basic or novel characteristics of that which is recited is not changed by the presence of more than that which is recited, but excludes prior art embodiments.
By "detectable amino acid sequence" or "detectable moiety" is meant a composition that when linked with the nucleic acid or protein molecule of interest renders the latter detectable, via any means, including spectroscopic, photochemical, biochemical, immunochemical, or chemical means. For example, useful labels include radioactive isotopes, magnetic beads, metallic beads, colloidal particles, fluorescent dyes, electron-dense reagents, enzymes (for example, as commonly used in an ELISA), biotin, digoxigenin, or haptens.
A "labeled nucleic acid or oligonucleotide probe" is one that is bound, either covalently, through a linker or a chemical bond, or noncovalently, through ionic bonds, van der Waals forces, electrostatic attractions, hydrophobic interactions, or hydrogen bonds, to a label such that the presence of the nucleic acid or probe may be detected by detecting the presence of the label bound to the nucleic acid or probe.
An "expression vector" is a nucleic acid construct, generated recombinantly or synthetically, bearing a series of specified nucleic acid elements that enable transcription of a particular gene in a host cell. Typically, gene expression is placed under the control of certain regulatory elements, including constitutive or inducible promoters, tissue-preferred regulatory elements, and enhancers.
By "an effective amount" is meant the amount of a required to ameliorate the symptoms of a disease relative to an untreated patient. The effective amount of active compound(s) used to practice the present invention for therapeutic treatment of a neoplastic disease varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the attending physician or veterinarian will decide the appropriate amount and dosage regimen. Such amount is referred to as an "effective" amount. By "fragment" is meant a portion (e.g., at least 10, 25, 50, 100, 125, 150, 200, 250,
300, 350, 400, or 500 amino acids or nucleic acids) of a protein or nucleic acid molecule that is substantially identical to a reference protein or nucleic acid and retains the biological activity of the reference. In some embodiments the portion retains at least 50%, 75%, or
80%, or more preferably 90%, 95%, or even 99% of the biological activity of the reference protein or nucleic acid described herein.
A "host cell" is any prokaryotic or eukaryotic cell that contains either a cloning vector or an expression vector. This term also includes those prokaryotic or eukaryotic cells that have been genetically engineered to contain the cloned gene(s) in the chromosome or genome of the host cell.
By "inhibitory nucleic acid" is meant a double-stranded RNA, siRNA (short interfering RNA), shRNA (short hairpin RNA), or antisense RNA, or a portion thereof, or a mimetic thereof, that when administered to a mammalian cell results in a decrease (e.g., by 10%, 25%, 50%, 75%, or even 90-100%) in the expression of a target gene. Typically, a nucleic acid inhibitor comprises at least a portion of a target nucleic acid molecule, or an ortholog thereof, or comprises at least a portion of the complementary strand of a target nucleic acid molecule.
The terms "isolated," "purified," or "biologically pure" refer to material that is free to varying degrees from components which normally accompany it as found in its native state. Various levels of purity may be applied as needed according to this invention in the different methodologies set forth herein; the customary purity standards known in the art may be used if no standard is otherwise specified.
By "isolated nucleic acid molecule" is meant a nucleic acid (e.g., a DNA, RNA, or analog thereof) that is free of the genes which, in the naturally-occurring genome of the organism from which the nucleic acid molecule of the invention is derived, flank the gene. The term therefore includes, for example, a recombinant DNA that is incorporated into a vector; into an autonomously replicating plasmid or virus; or into the genomic DNA of a prokaryote or eukaryote; or that exists as a separate molecule (for example, a cDNA or a genomic or cDNA fragment produced by PCR or restriction endonuclease digestion) independent of other sequences. In addition, the term includes an RNA molecule which is transcribed from a DNA molecule, as well as a recombinant DNA which is part of a hybrid gene encoding additional polypeptide sequence.
"Microarray" means a collection of nucleic acid molecules or polypeptides from one or more organisms arranged on a solid support (for example, a chip, plate, or bead).
By "neoplasia" is meant any disease that is caused by or results in inappropriately high levels of cell division, inappropriately low levels of apoptosis, or both. For example, cancer is an example of a neoplasia. Examples of cancers include, without limitation, leukemias (e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia,
acute myeloblasts leukemia, acute promyelocyte leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelocytic leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphoma (Hodgkin's disease, non-Hodgkin's disease), Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, nile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, uterine cancer, testicular cancer, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodenroglioma, schwannoma, meningioma, melanoma, neuroblastoma, and retinoblastoma). Lymphoproliferative disorders are also considered to be proliferative diseases.
By "nucleic acid" is meant an oligomer or polymer of ribonucleic acid or deoxyribonucleic acid, or analog thereof. This term includes oligomers consisting of naturally occurring bases, sugars, and intersugar (backbone) linkages as well as oligomers having non-naturally occurring portions which function similarly. Such modified or substituted oligonucleotides are often preferred over native forms because of properties such as, for example, enhanced stability in the presence of nucleases. Specific examples of some nucleic acids envisioned for this invention may contain phosphorothioates, phosphotriesters, methyl phosphonates, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar linkages. Also preferred are oligonucleotides having morpholino backbone structures (Summerton, J. E. and Weller, D. D., U.S. Pat. No: 5,034,506). In other preferred embodiments, such as the protein- nucleic acid (PNA) backbone, the phosphodiester backbone of the oligonucleotide may be replaced with a polyamide backbone, the bases being bound directly or indirectly to the aza nitrogen atoms of the polyamide backbone (P. E. Nielsen et al. Science 199: 254, 1997). Other preferred oligonucleotides may contain alkyl and halogen-substituted sugar moieties
comprising one of the following at the 2' position: OH, SH, SCH3, F5 OCN, O(CH2)nNH2 or O(CH2)n CH3, where n is from 1 to about 10; Ci to Cio lower alkyl, substituted lower alkyl, alkaryl or aralkyl; Cl; Br; CN; CF3; OCF3; O~, S-, or N-alkyl; O-, S-, or N-alkenyl; SOCH3; SO2CH3; ONO2; NO2; N3; NH2; heterocycloalkyl; heterocycloalkaryl; aminoalkylamino; polyalkylamino; substituted silyl; an RNA cleaving group; a conjugate; a reporter group; an intercalator; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide and other substituents having similar properties. Oligonucleotides may also have sugar mimetics such as cyclobutyls in place of the pentofuranosyl group. Other preferred embodiments may include at least one modified base form. Some specific examples of such modified bases include 2-(amino)adenine, 2-(methylamino)adenine, 2- (imidazolylalkyl)adenine, 2-(aminoalklyamino)adenine, or other heterosubstituted alkyladenines.
By "operably linked" is meant that a first polynucleotide is positioned adjacent to a second polynucleotide that directs transcription of the first polynucleotide when appropriate molecules (e.g., transcriptional activator proteins) are bound to the second polynucleotide. By "ortholog" is meant any protein or nucleic acid molecule of an organism that is highly related to a reference protein or nucleic acid sequence from another organism.
By "recombinant" is meant the product of genetic engineering or chemical synthesis. By "protein" is meant any chain of amino acids, or analogs thereof, regardless of length or post-translational modification.
By "positioned for expression" is meant that the polynucleotide of the invention (e.g., a DNA molecule) is positioned adjacent to a DNA sequence that directs transcription and translation of the sequence (i.e., facilitates the production of, for example, a recombinant protein of the invention, or an RNA molecule).
By "reference" is meant a standard or control condition.
By "siRNA" is meant a double stranded RNA. Optimally, an siRNA is 18, 19, 20, 21, 22, 23 or 24 nucleotides in length and has a 2 base overhang at its 3' end. These dsRNAs can be introduced to an individual cell or to a whole animal; for example, they may be introduced systemically via the bloodstream. Such siRNAs are used to downregulate mRNA levels or promoter activity.
By "specifically binds" is meant a molecule (e.g., peptide, polynucleotide) that recognizes and binds a protein or nucleic acid molecule of the invention, but which does not
substantially recognize and bind other molecules in a sample, for example, a biological sample, which naturally includes a protein of the invention.
By "subject" is meant a mammal, including, but not limited to, a human or non-human mammal, such as a bovine, equine, canine, ovine, or feline. By "substantially identical" is meant a protein or nucleic acid molecule exhibiting at least 50% identity to a reference amino acid sequence (for example, any one of the amino acid sequences described herein) or nucleic acid sequence (for example, any one of the nucleic acid sequences described herein). Preferably, such a sequence is at least 60%, more preferably 80% or 85%, and most preferably 90%, 95% or even 99% identical at the amino acid level or nucleic acid to the sequence used for comparison.
Sequence identity is typically measured using sequence analysis software (for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs). Such software matches identical or similar sequences by assigning degrees of homology to various substitutions, deletions, and/or other modifications. Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. In an exemplary approach to determining the degree of identity, a BLAST program may be used, with a probability score between e'3 and e~100 indicating a closely related sequence.
By "transformed cell" is meant a cell into which (or into an ancestor of which) has been introduced, by means of recombinant DNA techniques, a polynucleotide molecule encoding (as used herein) a protein of the invention.
Methods of the Invention
The invention features compositions and methods useful for the diagnosis of a neoplasia in a subject. These methods and compositions are based, in part, on the discovery that ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3) expression is increased in neoplastic tissues. In addition, the invention also provides methods and compositions for altering ASPM expression in a neoplastic cell. Such compositions and methods are likely to be useful for the treatment of neoplasia.
Diagnostics
Neoplastic tissues express higher levels of an ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3) or an ASPM polynucleotide than corresponding normal tissues. Accordingly, expression levels of an ASPM nucleic acid molecule or polypeptide are correlated with a particular disease state (e.g., neoplasia), and thus are useful in diagnosis. Accordingly, the present invention provides a number of diagnostic assays that are useful for the identification or characterization of a neoplasia.
In one embodiment, a patient having a neoplasia will show an increase in the expression of an ASPM nucleic acid molecule. Alterations in gene expression are detected using methods known to the skilled artisan and described herein. Such information can be used to diagnose a neoplasia. In another embodiment, an alteration in the expression of an ASPM nucleic acid molecule is detected using real-time quantitative PCR (Q-rt-PCR) to detect changes in gene expression.
Primers used for amplification of an ASPM nucleic acid molecule, including but not limited to those primer sequences described herein, are useful in diagnostic methods of the invention. The primers of the invention embrace oligonucleotides of sufficient length and appropriate sequence so as to provide specific initiation of polymerization on a significant number of nucleic acids. Specifically, the term "primer" as used herein refers to a sequence comprising two or more deoxyribonucleotides or ribonucleotides, preferably more than three, and most preferably more than 8, which sequence is capable of initiating synthesis of a primer extension product, which is substantially complementary to a locus strand. The primer must be sufficiently long to prime the synthesis of extension products in the presence of the inducing agent for polymerization. The exact length of primer will depend on many factors, including temperature, buffer, and nucleotide composition. The oligonucleotide primer typically contains between 12 and 27 or more nucleotides, although it may contain fewer nucleotides. Primers of the invention are designed to be "substantially" complementary to each strand of the genomic locus to be amplified and include the appropriate G or C nucleotides as discussed above. This means that the primers must be sufficiently complementary to hybridize with their respective strands under conditions that allow the agent for polymerization to perform. In other words, the primers should have sufficient complementarity with the 5' and 3' flanking sequences to hybridize therewith and permit amplification of the genomic locus. While exemplary primers are provided herein, it is understood that any primer that hybridizes with the target sequences of the invention are useful in the method of the invention for detecting ASPM nucleic acid molecules.
In one embodiment, ASPM-specific primers amplify a desired genomic target using the polymerase chain reaction (PCR). The amplified product is then detected using standard methods known in the art. In one embodiment, a PCR product (i.e., amplicon) or real-time PCR product is detected by probe binding. In one embodiment, probe binding generates a fluorescent signal, for example, by coupling a fluorogenic dye molecule and a quencher moiety to the same or different oligonucleotide substrates (e.g., TaqMan® (Applied Biosystems, Foster City, CA, USA), Molecular Beacons (see, for example, Tyagi et al., Nature Biotechnology 14(3):303-8, 1996), Scorpions® (Molecular Probes Inc., Eugene, OR, USA)). In another example, a PCR product is detected by the binding of a fluorogenic dye that emits a fluorescent signal upon binding (e.g., SYBR® Green (Molecular Probes)). Such detection methods are useful for the detection of an ASPM PCR product. ,
In another embodiment, hybridization with PCR probes that are capable of detecting an ASPM nucleic acid molecule, including genomic sequences, or closely related molecules, may be used to hybridize to a nucleic acid sequence derived from a patient having a neoplasia. The specificity of the probe determines whether the probe hybridizes to a naturally occurring sequence, allelic variants, or other related sequences. Hybridization techniques may be used to identify mutations indicative of a neoplasia, or may be used to monitor expression levels of these genes (for example, by Northern analysis (Ausubel et al., supra). In yet another embodiment, humans may be diagnosed for a propensity to develop a neoplasia by direct analysis of the sequence of an ASPM nucleic acid molecule. The sequence of an ASPM nucleic acid molecule derived from a subject is compared to a reference sequence. An alteration in the sequence of the ASPM nucleic acid molecule relative to the reference indicates that the patient has or has a propensity to develop a neoplasia.
In another approach, diagnostic methods of the invention are used to assay the expression of an ASPM polypeptide in a biological sample relative to a reference (e.g., the level of ASPM polypeptide present in a corresponding control tissue). In one embodiment, the level of an ASPM polypeptide is detected using an antibody that specifically binds an ASPM polypeptide. Exemplary antibodies that specifically bind an ASPM polypeptide are described herein. Such antibodies are useful for the diagnosis of a neoplasia. Methods for measuring an antibody- ASPM complex include, for example, detection of fluorescence, luminescence, chemiluminescence, absorbance, reflectance, transmittance, birefringence or refractive index. Optical methods include microscopy (both confocal and non-confocal),
imaging methods and non-imaging methods. Methods for performing these assays are readily known in the art. Useful assays include, for example, an enzyme immune assay (EIA) such as enzyme-linked immunosorbent assay (ELISA), a radioimmune assay (RIA), a Western blot assay, or a slot blot assay. These methods are also described in, e.g., Methods in Cell Biology: Antibodies in Cell Biology, volume 37 (Asai, ed. 1993); Basic and Clinical Immunology (Stites & Terr, eds., 7th ed. 1991); and Harlow & Lane, supra. Immunoassays can be used to determine the quantity of ASPM in a sample, where an increase in the level of the ASPM polypeptide is diagnostic of a patient having a neoplasia.
In general, the measurement of an ASPM polypeptide or nucleic acid molecule in a subject sample is compared with a diagnostic amount present in a reference. A diagnostic amount distinguishes between a neoplastic tissue and a control tissue. The skilled artisan appreciates that the particular diagnostic amount used can be adjusted to increase sensitivity or specificity of the diagnostic assay depending on the preference of the diagnostician. In general, any significant increase (e.g., at least about 10%, 15%, 30%, 50%, 60%, 75%, 80%, or 90%) in the level of an ASPM polypeptide or nucleic acid molecule in the subject sample relative to a reference may be used to diagnose a neoplasia. In one embodiment, the reference is the level of ASPM polypeptide or nucleic acid molecule present in a control sample obtained from a patient that does not have a neoplasia. In another embodiment, the reference is a baseline level of ASPM present in a biologic sample derived from a patient prior to, during, or after treatment for a neoplasia. In yet another embodiment, the reference is a standardized curve.
Types of Biological Samples
The level of an ASPM polypeptide or nucleic acid molecule can be measured in different types of biologic samples. In one embodiment, the biologic sample is a tissue sample that includes cells of a tissue or organ. Such tissue is obtained, for example, from a biopsy. In another embodiment, the biologic sample is a biologic fluid sample (e.g., blood, blood plasma, serum, urine, seminal fluids, ascites, or cerebrospinal fluid).
Kits
The invention also provides kits for the diagnosis or monitoring of a neoplasia in a biological sample obtained from a subject. In one embodiment, the kit detects an increase in the expression of an ASPM nucleic acid molecule or polypeptide relative to a reference level of expression. In another embodiment, the kit detects an alteration in the sequence of an
ASPM nucleic acid molecule derived from a subject relative to a reference sequence. In related embodiments, the kit includes reagents for monitoring the expression of an ASPM nucleic acid molecule, such as primers or probes that hybridize to an ASPM nucleic acid molecule. In other embodiments, the kit includes an antibody that binds to an ASPM polypeptide.
Optionally, the kit includes directions for monitoring ASPM nucleic acid molecule or polypeptide levels in a biological sample derived from a subject. In other embodiments, the kit comprises a sterile container which contains the primer, probe, antibody, or other detection regents; such containers can be boxes, ampules, bottles, vials, tubes, bags, pouches, blister-packs, or other suitable container form known in the art. Such containers can be made of plastic, glass, laminated paper, metal foil, or other materials suitable for holding nucleic acids. The instructions will generally include information about the use of the primers or probes described herein and their use in diagnosing a neoplasia. Preferably, the kit further comprises any one or more of the reagents described in the diagnostic assays described herein. In other embodiments, the instructions include at least one of the following: description of the primer or probe; methods for using the enclosed materials for the diagnosis of a neoplasia; precautions; warnings; indications; clinical or research studies; and/or references. The instructions may be printed directly on the container (when present), or as a label applied to the container, or as a separate sheet, pamphlet, card, or folder supplied in or with the container.
Patient Monitoring
The disease state or treatment of a patient having a neoplasia can be monitored using the methods and compositions of the invention. In one embodiment, a microarray is used to assay the expression profile of an ASPM nucleic acid molecule. Such monitoring may be useful, for example, in assessing the efficacy of a particular drug in a patient. Therapeutics that alter the expression of an ASPM nucleic acid molecule or ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3), are taken as particularly useful in the invention.
ASPM Antibodies
Antibodies are well known to those of ordinary skill in the science of immunology. As used herein, the term "antibody" means not only intact antibody molecules, but also fragments of antibody molecules that retain immunogen binding ability. Such fragments are also well known in the art and are regularly employed both in vitro and in vivo. Accordingly,
as used herein, the term "antibody" means not only intact immunoglobulin molecules but also the well-known active fragments F(ab')2, and Fab. F(ab')2, and Fab fragments which lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding of an intact antibody (Wahl et al., J. Nucl. Med. 24:316-325 (1983). The antibodies of the invention comprise whole native antibodies, bispecific antibodies; chimeric antibodies; Fab, Fab', single chain V region fragments (scFv) and fusion polypeptides.
In one embodiment, an antibody that binds an ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3) is monoclonal. Alternatively, the anti-ASPM antibody is a polyclonal antibody. The preparation and use of polyclonal antibodies are also known the skilled artisan. The invention also encompasses hybrid antibodies, in which one pair of heavy and light chains is obtained from a first antibody, while the other pair of heavy and light chains is obtained from a different second antibody. Such hybrids may also be formed using humanized heavy and light chains. Such antibodies are often referred to as "chimeric" antibodies.
In general, intact antibodies are said to contain "Fc" and "Fab" regions. The Fc regions are involved in complement activation and are not involved in antigen binding. An antibody from which the Fc' region has been enzymatically cleaved, or which has been produced without the Fc' region, designated an "F(ab')2" fragment, retains both of the antigen binding sites of the intact antibody. Similarly, an antibody from which the Fc region has been enzymatically cleaved, or which has been produced without the Fc region, designated an "Fab"' fragment, retains one of the antigen binding sites of the intact antibody. Fab' fragments consist of a covalently bound antibody light chain and a portion of the antibody heavy chain, denoted "Fd." The Fd fragments are the major determinants of antibody specificity (a single Fd fragment may be associated with up to ten different light chains without altering antibody specificity). Isolated Fd fragments retain the ability to specifically bind to immunogenic epitopes.
Antibodies can be made by any of the methods known in the art utilizing ASPM polypeptides (e.g., ASPM, ASPM variant 1, 2, or 3), or immunogenic fragments thereof, as an immunogen. One method of obtaining antibodies is to immunize suitable host animals with an immunogen and to follow standard procedures for polyclonal or monoclonal antibody production. The immunogen will facilitate presentation of the immunogen on the cell surface. Immunization of a suitable host can be carried out in a number of ways. Nucleic
acid sequences encoding an ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3), or immunogenic fragments thereof, can be provided to the host in a delivery vehicle that is taken up by immune cells of the host. The cells will in turn express the receptor on the cell surface generating an immunogenic response in the host. Alternatively, nucleic acid sequences encoding an ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3), or immunogenic fragments thereof, can be expressed in cells in vitro, followed by isolation of the receptor and administration of the receptor to a suitable host in which antibodies are raised.
Using either approach, antibodies can then be purified from the host. Antibody purification methods may include salt precipitation (for example, with ammonium sulfate), ion exchange chromatography (for example, on a cationic or anionic exchange column preferably run at neutral pH and eluted with step gradients of increasing ionic strength), gel filtration chromatography (including gel filtration HPLC), and chromatography on affinity resins such as protein A, protein G, hydroxyapatite, and antiimmunoglobulin.
Antibodies can be conveniently produced from hybridoma cells engineered to express the antibody. Methods ofmaking hybridomas are well known in the art. The hybridoma cells can be cultured in a suitable medium, and spent medium can be used as an antibody source. Polynucleotides encoding the antibody of interest can in turn be obtained from the hybridoma that produces the antibody, and then the antibody may be produced synthetically or recombinantly from these DNA sequences. For the production of large amounts of antibody, it is generally more convenient to obtain an ascites fluid. The method of raising ascites generally comprises injecting hybridoma cells into an immunologically naive histocompatible or immunotolerant mammal, especially a mouse. The mammal may be primed for ascites production by prior administration of a suitable composition; e.g., Pristane. Monoclonal antibodies (Mabs) produced by methods of the invention can be "humanized" by methods known in the art. "Humanized" antibodies are antibodies in which at least part of the sequence has been altered from its initial form to render it more like human immunoglobulins. Techniques to humanize antibodies are particularly useful when non- human animal (e.g., murine) antibodies are generated. Examples of methods for humanizing a murine antibody are provided in U.S. Patent Nos. 4,816,567, 5,530,101, 5,225,539, 5,585,089, 5,693,762 and 5,859,205.
ASPM Polypeptide Expression
In general, ASPM polypeptides, variants, and fragments thereof may be produced by transformation of a suitable host cell with all or part of a polypeptide-encoding nucleic acid molecule or fragment thereof in a suitable expression vehicle. Those skilled in the field of molecular biology will understand that any of a wide variety of expression systems may be used to provide the recombinant protein. The precise host cell used is not critical to the invention. A polypeptide of the invention may be produced in a prokaryotic host (e.g., E. coli) or in a eukaryotic host (e.g., Saccharomyces cerevisiae, insect cells, e.g., S£21 cells, or mammalian cells, e.g., NIH 3T3, HeLa, or preferably COS cells). Such cells are available from a wide range of sources (e.g., the American Type Culture Collection, Rockland, Md.; also, see, e.g., Ausubel et al., supra). The method of transformation or transfection and the choice of expression vehicle will depend on the host system selected. Transformation and transfection methods are described, e.g., in Ausubel et al. (supra); expression vehicles may be chosen from those provided, e.g., in Cloning Vectors: A Laboratory Manual (P. H. Pouwels et al., 1985, Supp. 1987).
A variety of expression systems exist for the production of the polypeptides of the invention. Expression vectors useful for producing such polypeptides include, without limitation, chromosomal, episomal, and virus-derived vectors, e.g., vectors derived from bacterial plasmids, from bacteriophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, and vectors derived from combinations thereof.
One particular bacterial expression system for polypeptide production is the E. coli pET expression system (Novagen, Inc., Madison, Wis). According to this expression system, DNA encoding a polypeptide is inserted into a pET vector in an orientation designed to allow expression. Since the gene encoding such a polypeptide is under the control of the T7 regulatory signals, expression of the polypeptide is achieved by inducing the expression of T7 RNA polymerase in the host cell. This is typically achieved using host strains that express T7 RNA polymerase in response to IPTG induction. Once produced, recombinant polypeptide is then isolated according to standard methods known in the art, for example, those described herein.
Another bacterial expression system for polypeptide production is the pGEX expression system (Pharmacia). This system employs a GST gene fusion system that is designed for high-level expression of genes or gene fragments as fusion proteins with rapid
purification and recovery of functional gene products. The protein of interest is fused to the carboxyl terminus of the glutathione S-transferase protein from Schistosoma japonicum and is readily purified from bacterial lysates by affinity chromatography using Glutathione Sepharose 4B. Fusion proteins can be recovered under mild conditions by elution with glutathione. Cleavage of the glutathione S-transferase domain from the fusion protein is facilitated by the presence of recognition sites for site-specific proteases upstream of this domain. For example, proteins expressed in pGEX-2T plasmids may be cleaved with thrombin; those expressed in pGEX-3X may be cleaved with factor Xa.
Once the recombinant polypeptide of the invention is expressed, it is isolated, e.g., using affinity chromatography. In one example, an antibody (e.g., produced as described herein) raised against a polypeptide of the invention may be attached to a column and used to isolate the recombinant polypeptide. Lysis and fractionation of polypeptide-harboring cells prior to affinity chromatography may be performed by standard methods (see, e.g., Ausubel et al., supra). Once isolated, the recombinant protein can, if desired, be further purified, e.g., by high performance liquid chromatography (see, e.g., Fisher, Laboratory Techniques In Biochemistry and Molecular Biology, eds., Work and Burdon, Elsevier, 1980). Polypeptides of the invention, particularly short peptide fragments, can also be produced by chemical synthesis (e.g., by the methods described in Solid Phase Peptide Synthesis, 2nd ed., 1984 The Pierce Chemical Co., Rockford, 111.). These general techniques of polypeptide expression and purification can also be used to produce and isolate useful peptide fragments or analogs (described herein).
ASPM Polypeptides and Analogs Also included in the invention are ASPM polypeptides, variants, or fragments thereof containing at least one alteration relative to a reference sequence. Such alterations include certain mutations, deletions, insertions, or post-translational modifications. The invention further includes analogs of any naturally-occurring polypeptide of the invention. Analogs can differ from naturally-occurring polypeptides of the invention by amino acid sequence differences, by post-translational modifications, or by both. Analogs of the invention will generally exhibit at least 85%, more preferably 90%, and most preferably 95% or even 99% identity with all or part of a naturally-occurring amino acid sequence of the invention. The length of sequence comparison is at least 10, 13, 15 amino acid residues, preferably at least 25 amino acid residues, and more preferably more than 35 amino acid residues. Again, in an
exemplary approach to determining the degree of identity, a BLAST program may be used, with a probability score between e"3 and e"100 indicating a closely related sequence. Modifications include in vivo and in vitro chemical derivatization of polypeptides, e.g., acetylation, carboxylation, phosphorylation, or glycosylation; such modifications may occur during polypeptide synthesis or processing or following treatment with isolated modifying enzymes. Analogs can also differ from the naturally-occurring polypeptides of the invention by alterations in primary sequence. These include genetic variants, both natural and induced (for example, resulting from random mutagenesis by irradiation or exposure to ethanemethylsulfate or by site-specific mutagenesis as described in Sambrook, Fritsch and Maniatis, Molecular Cloning: A Laboratory Manual (2d ed.), CSH Press, 1989, or Ausubel et al., supra). Also included are cyclized peptides, molecules, and analogs which contain residues other than L-amino acids, e.g., D-amino acids or non-naturally occurring or synthetic amino acids.
In addition to full-length polypeptides, the invention also includes fragments of any one of the polypeptides of the invention. As used herein, the term "a fragment" means at least 5, 10, 13, or 15 amino acids. In other embodiments a fragment is at least 20 contiguous amino acids, at least 30 contiguous amino acids, or at least 50 contiguous amino acids, and in other embodiments at least 60 to 80 or more contiguous amino acids. Fragments of the invention can be generated by methods known to those skilled in the art or may result from normal protein processing (e.g., removal of amino acids from the nascent polypeptide that are not required for biological activity or removal of amino acids by alternative mRNA splicing or alternative protein processing events).
ASPM Polynucleotides In general, the invention includes any nucleic acid sequence encoding an ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3). Also included in the methods of the invention are any nucleic acid molecule containing at least one strand that hybridizes with such a nucleic acid sequence (e.g., an inhibitory nucleic acid molecule, such as a dsRNA, siRNA, shRNA, or antisense molecule). An isolated nucleic acid molecule can be manipulated using recombinant DNA techniques well known in the art. Thus, a nucleotide sequence contained in a vector in which 5' and 3' restriction sites are known, or for which polymerase chain reaction (PCR) primer sequences have been disclosed, is considered isolated, but a nucleic acid sequence existing in its native state in its natural host is not. An isolated nucleic acid may be substantially purified, but need not be. For example, a nucleic
acid molecule that is isolated within a cloning or expression vector may comprise only a tiny percentage of the material in the cell in which it resides. Such a nucleic acid is isolated, however, as the term is used herein, because it can be manipulated using standard techniques known to those of ordinary skill in the art.
ASPM Polynucleotide Therapy
Polynucleotide therapy featuring a polynucleotide encoding an ASPM protein, variant, or fragment thereof is another therapeutic approach for treating a neoplasia. Such nucleic acid molecules can be delivered to cells of a subject having a neoplasia. The nucleic acid molecules must be delivered to the cells of a subject in a form in which they can be taken up so that therapeutically effective levels of an ASPM protein (e.g., ASPM, ASPM variant 1, 2, or 3) or fragment thereof can be produced.
Transducing viral (e.g., retroviral, adenoviral, and adeno-associated viral) vectors can be used for somatic cell gene therapy, especially because of their high efficiency of infection and stable integration and expression (see, e.g., Cayouette et al., Human Gene Therapy 8:423-430, 1997; Kido et al., Current Eye Research 15:833-844, 1996; Bloomer et al., Journal of Virology 71 :6641-6649, 1997; Naldini et al., Science 272:263-267, 1996; and Miyoshi et al., Proc. Natl. Acad. Sci. U.S.A. 94:10319, 1997). For example, a polynucleotide encoding an ASPM protein, variant, or a fragment thereof, can be cloned into a retroviral vector and expression can be driven from its endogenous promoter, from the retroviral long terminal repeat, or from a promoter specific for a target cell type of interest. Other viral vectors that can be used include, for example, a vaccinia virus, a bovine papilloma virus, or a herpes virus, such as Epstein-Barr Virus (also see, for example, the vectors of Miller, Human Gene Therapy 15-14, 1990; Friedman, Science 244:1275-1281, 1989; Eglitis et al., BioTechniques 6:608-614, 1988; Tolstoshev et al., Current Opinion in Biotechnology 1:55- 61, 1990; Sharp, The Lancet 337:1277-1278, 1991; Cornetta et al., Nucleic Acid Research and Molecular Biology 36:311-322, 1987; Anderson, Science 226:401-409, 1984; Moen, Blood Cells 17:407-416, 1991; Miller et al., Biotechnology 7:980-990, 1989; Le Gal La Salle et al., Science 259:988-990, 1993; and Johnson, Chest 107:77S-83S, 1995). Retroviral vectors are particularly well developed and have been used in clinical settings (Rosenberg et al., N. Engl. J. Med 323:370, 1990; Anderson et al., U.S. Pat. No. 5,399,346). Most preferably, a viral vector is used to administer an ASPM polynucleotide systemically.
Non- viral approaches can also be employed for the introduction of therapeutic to a cell of a patient diagnosed as having a neoplasia. For example, a nucleic acid molecule can
be introduced into a cell by administering the nucleic acid in the presence of lipofection (Feigner et al., Proc. Natl. Acad. Sci. U.S.A. 84:7413, 1987; Ono et al., Neuroscience Letters 17:259, 1990; Brigham et al., Am. J. Med. Sci. 298:278, 1989; Staubinger et al., Methods in Enzymology 101:512, 1983), asialoorosomucoid-polylysine conjugation (Wu et al., Journal of Biological Chemistry 263:14621, 1988; Wu et al., Journal of Biological Chemistry 264:16985, 1989), or by micro-injection under surgical conditions (Wolff et al., Science 247: 1465, 1990). Preferably the nucleic acids are administered in combination with a liposome and protamine.
Gene transfer can also be achieved using non-viral means involving transfection in vitro. Such methods include the use of calcium phosphate, DEAE dextran, electroporation, and protoplast fusion. Liposomes can also be potentially beneficial for delivery of DNA into a cell. Transplantation of normal genes into the affected tissues of a patient can also be accomplished by transferring a normal nucleic acid into a cultivatable cell type ex vivo (e.g., an autologous or heterologous primary cell or progeny thereof), after which the cell (or its descendants) are injected into a targeted tissue. cDNA expression for use in polynucleotide therapy methods can be directed from any suitable promoter (e.g., the human cytomegalovirus (CMV), simian virus 40 (SV40), or metallothionein promoters), and regulated by any appropriate mammalian regulatory element. For example, if desired, enhancers known to preferentially direct gene expression in specific cell types can be used to direct the expression of a nucleic acid. The enhancers used can include, without limitation, those that are characterized as tissue- or cell-specific enhancers. Alternatively, if a genomic clone is used as a therapeutic construct, regulation can be mediated by the cognate regulatory sequences or, if desired, by regulatory sequences derived from a heterologous source, including any of the promoters or regulatory elements described above.
Another therapeutic approach included in the invention involves administration of a recombinant therapeutic, such as a recombinant ASPM protein, variant, or fragment thereof, either directly to the site of a potential or actual disease-affected tissue or systemically (for example, by any conventional recombinant protein administration technique). The dosage of the administered protein depends on a number of factors, including the size and health of the individual patient. For any particular subject, the specific dosage regimes should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions.
Screening Assays
As reported herein, the expression of an ASPM polypeptide is increased in neoplastic tissues. Accordingly, compounds that modulate the expression or activity of an ASPM polypeptide, variant, or fragment thereof are useful in the methods of the invention for the treatment or prevention of a neoplasm (e.g., breast, colon, lymph, ovary, stomach, thyroid, testis, and uterine cancer). Any number of methods are available for carrying out screening assays to identify such compounds. In one approach, candidate compounds are identified that specifically bind to and alter the activity of a polypeptide of the invention (e.g., an ASPM activity associated with cell proliferation, mitosis or maintenance of chromosomal stability). Methods of assaying such biological activities are known in the art and are described herein. The efficacy of such a candidate compound is dependent upon its ability to interact with an ASPM polypeptide, variant, or fragment. Such an interaction can be readily assayed using any number of standard binding techniques and functional assays (e.g., those described in Ausubel et al., supra). For example, a candidate compound may be tested in vitro for interaction and binding with a polypeptide of the invention and its ability to modulate cell proliferation, mitosis, or maintenance of chromosomal stability. ASPM's function in chromosomal stability can be assayed by detecting, for example, chromosomal nondysjunction in cells where endogenous ASPM expression is perturbed or reduced. Standard methods for perturbing or reducing ASPM expression include mutating or deleting an endogenous ASPM sequence, interfering with ASPM expression using RNAi, or microinjecting an ASPM-expressing cell with an antibody that binds ASPM and interferes with its function. Alternatively, chromosomal nondysjunction can be assayed in vivo, for example, in a mouse model in which ASPM has been knocked out by homologous recombination, or any other standard method. Potential agonists and antagonists of an ASPM polypeptide include organic molecules, peptides, peptide mimetics, polypeptides, nucleic acid molecules (e.g., double- stranded RNAs, siRNAs, antisense polynucleotides), and antibodies that bind to a nucleic acid sequence or polypeptide of the invention and thereby inhibit or extinguish its activity. Potential antagonists also include small molecules that bind to the ASPM polypeptide thereby preventing binding to cellular molecules with which the ASPM polypeptide normally interacts, such that the normal biological activity of the ASPM polypeptide is reduced or inhibited. Small molecules of the invention preferably have a molecular weight below 2,000 daltons, more preferably between 300 and 1,000 daltons, and most preferably between 400 and 700 daltons. It is preferred that these small molecules are organic molecules.
In one particular example, a candidate compound that binds to an ASPM polypeptide, variant, or fragment thereof may be identified using a chromatography-based technique. For example, a recombinant polypeptide of the invention may be purified by standard techniques from cells engineered to express the polypeptide (e.g., those described above) and may be immobilized on a column. A solution of candidate compounds is then passed through the column, and a compound specific for the ASPM polypeptide is identified on the basis of its ability to bind to the ASPM polypeptide and be immobilized on the column. To isolate the compound, the column is washed to remove non-specifically bound molecules, and the compound of interest is then released from the column and collected. Similar methods may be used to isolate a compound bound to a polypeptide microarray. Compounds isolated by this method (or any other appropriate method) may, if desired, be further purified (e.g., by high performance liquid chromatography). In addition, these candidate compounds may be tested for their ability to alter the biological activity of an ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3). Compounds that are identified as binding to a polypeptide of the invention with an affinity constant less than or equal to 10 mM are considered particularly useful in the invention. Alternatively, any in vivo protein interaction detection system, for example, any two-hybrid assay may be utilized to identify compounds that interact with an ASPM polypeptide. Interacting compounds isolated by this method (or any other appropriate , method) may, if desired, be further purified (e.g., by high performance liquid chromatography). Compounds isolated by any approach described herein may be used as therapeutics to treat a neoplasia in a human patient.
In addition, compounds that inhibit the expression of an ASPM nucleic acid molecule whose expression is increased in a patient having a neoplasia are also useful in the methods of the invention. Any number of methods are available for carrying out screening assays to identify new candidate compounds that alter the expression of an ASPM nucleic acid molecule. In one working example, candidate compounds are added at varying concentrations to the culture medium of cultured cells expressing one of the nucleic acid sequences of the invention. Gene expression is then measured, for example, by microarray analysis, Northern blot analysis (Ausubel et al., supra), or RT-PCR, using any appropriate fragment prepared from the nucleic acid molecule as a hybridization probe. The level of gene expression in the presence of the candidate compound is compared to the level measured in a control culture medium lacking the candidate molecule. A compound that promotes an alteration in the expression of an ASPM gene, or a functional equivalent thereof, is
considered useful in the invention; such a molecule may be used, for example, as a therapeutic to treat a neoplasia in a human patient.
In another approach, the effect of candidate compounds is measured at the level of polypeptide production to identify those that promote an alteration in an ASPM polypeptide level. The level of ASPM polypeptide can be assayed using any standard method. Standard immunological techniques include Western blotting or immunoprecipitation with an antibody specific for an ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3). For example, immunoassays may be used to detect or monitor the expression of at least one of the polypeptides of the invention in an organism. Polyclonal or monoclonal antibodies (produced as described above) that are capable of binding to such a polypeptide may be used in any standard immunoassay format (e.g., ELISA, Western blot, or RIA assay) to measure the level of the polypeptide. In some embodiments, a compound that promotes a decrease in the expression or biological activity of the polypeptide is considered particularly useful. Again, such a molecule may be used, for example, as a therapeutic to delay, ameliorate, or treat a neoplasia in a human patient.
In another embodiment, a nucleic acid described herein (e.g., an ASPM nucleic acid) is expressed as a transcriptional or translational fusion with a detectable reporter, and expressed in an isolated cell (e.g., mammalian or insect cell) under the control of a heterologous promoter, such as an inducible promoter. The cell expressing the fusion protein is then contacted with a candidate compound, and the expression of the detectable reporter in that cell is compared to the expression of the detectable reporter in an untreated control cell. A candidate compound that alters the expression of the detectable reporter is a compound that is useful for the treatment of a neoplasia. In one embodiment, the compound decreases the expression of the reporter. Each of the DNA sequences listed herein may also be used in the discovery and development of a therapeutic compound for the treatment of neoplasia. The encoded protein, upon expression, can be used as a target for the screening of drugs. Additionally, the DNA sequences encoding the amino terminal regions of the encoded protein or Shine-Delgarno or other translation facilitating sequences of the respective mRNA can be used to construct sequences that promote the expression of the coding sequence of interest. Such sequences may be isolated by standard techniques (Ausubel et al., supra).
The invention also includes novel compounds identified by the above-described screening assays. Optionally, such compounds are characterized in one or more appropriate animal models to determine the efficacy of the compound for the treatment of a neoplasia.
Desirably, characterization in an animal model can also be used to determine the toxicity, side effects, or mechanism of action of treatment with such a compound. Furthermore, novel compounds identified in any of the above-described screening assays may be used for the treatment of a neoplasia in a subject. Such compounds are useful alone or in combination with other conventional therapies known in the art.
Test Compounds and Extracts
In general, compounds capable of inhibiting the growth or proliferation of a neoplasia by altering the expression or biological activity of an ASPM polypeptide, variant, or fragment thereof are identified from large libraries of either natural product or synthetic (or semisynthetic) extracts or chemical libraries according to methods known in the art. Numerous methods are also available for generating random or directed synthesis (e.g., semi-synthesis or total synthesis) of any number of chemical compounds, including, but not limited to, saccharide-, lipid-, peptide-, and nucleic acid-based compounds. Synthetic compound libraries are commercially available from Brandon Associates (Merrimack, N.H.) and Aldrich Chemical (Milwaukee, Wis.). Alternatively, libraries of natural compounds in the form of " bacterial, fungal, plant, and animal extracts are commercially available from a number of sources, including Biotics (Sussex, UK), Xenova (Slough, UK), Harbor Branch Oceangraphics Institute (Ft. Pierce, FIa.), and PharmaMar, U.S.A. (Cambridge, Mass.). In one embodiment, test compounds of the invention are present in any combinatorial library known in the art, including: biological libraries; peptoid libraries (libraries of molecules having the functionalities of peptides, but with a novel, non-peptide backbone which are resistant to enzymatic degradation but which nevertheless remain bioactive; see, e.g., Zuckermann, R.N. et al, J. Med. Chem. 37:2678-85, 1994); spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the 'one-bead one-compound1 library method; and synthetic library methods using affinity chromatography selection. The biological library and peptoid library approaches are limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam, Anticancer DrugDes. 12:145, 1997).
Examples of methods for the synthesis of molecular libraries can be found in the art, for example in: DeWitt et al, Proc. Natl. Acad. ScL U.S.A. 90:6909, 1993; Erb et al, Proc. Natl. Acad. ScL USA 91:11422, 1994; Zuckermann et al, J. Med. Chem. 37:2678, 1994; Cho et al, Science 261:1303, 1993; Carrell et al, Angew. Chem. Int. Ed. Engl. 33:2059, 1994;
Carell et al, Angew. Chem. Int. Ed. Engl. 33:2061, 1994; and Gallop et al, J. Med. Client.
37:1233, 1994.
Libraries of compounds may be presented in solution (e.g., Houghten, Biotechniques
13:412-421, 1992), or on beads (Lam, Nature 354:82-84, 1991), chips (Fodor, Nature 364:555-556, 1993), bacteria (Ladner, U.S. Patent No. 5,223,409), spores (Ladner U.S. Patent
No. 5,223,409), plasmids (Cull et al, Proc Natl Acad Sd USA 89:1865-1869, 1992) or on phage (Scott and Smith, Science 249:386-390, 1990; Devlin, Science 249:404-406, 1990;
Cwirla et al. Proc. Natl. Acad. Sd. 87:6378-6382, 1990; Felici, J. MoI. Biol. 222:301-310,
1991; Ladner supra.). In addition, those skilled in the art of drug discovery and development readily understand that methods for dereplication (e.g., taxonomic dereplication, biological dereplication, and chemical dereplication, or any combination thereof) or the elimination of replicates or repeats of materials already known for their anti-neoplastic activity should be employed whenever possible. Those skilled in the field of drug discovery and development will understand that the precise source of a compound or test extract is not critical to the screening procedure(s) of the invention. Accordingly, virtually any number of chemical extracts or compounds can be screened using the methods described herein. Examples of such extracts or compounds include, but are not limited to, plant-, fungal-, prokaryotic- or animal-based extracts, fermentation broths, and synthetic compounds, as well as modification of existing compounds.
When a crude extract is found to alter the biological activity of an ASPM polypeptide, variant, or fragment thereof, further fractionation of the positive lead extract is necessary to isolate chemical constituents responsible for the observed effect. Thus, the goal of the extraction, fractionation, and purification process is the careful characterization and identification of a chemical entity within the crude extract having anti-neoplastic activity.
Methods of fractionation and purification of such heterogenous extracts are known in the art.
If desired, compounds shown to be useful agents for the treatment of a neoplasm are chemically modified according to methods known in the art.
Methods of Assaying ASPM Biological Activity
Therapeutics useful in the methods of the invention include, but are not limited to, those that have an anti-neoplastic activity or those that alter an ASPM biological activity associated with cell proliferation or maintenance of chromosomal stability. Neoplastic cell
growth is not subject to the same regulatory mechanisms that govern the growth or proliferation of normal cells. Compounds that reduce the growth or proliferation of a neoplasm are useful for the treatment of neoplasms. Methods of assaying cell growth and proliferation are known in the art. See, for example, Kittler et al. (Nature. 432 (7020): 1036- 40, 2004) and by Miyamoto et al. (Nature 416(6883):865-9, 2002). Assays for cell proliferation generally involve the measurement of DNA synthesis during cell replication. In one embodiment, DNA synthesis is detected using labeled DNA precursors, such as ([3H]- Thymidine or 5-bromo-2'-deoxyuridine [BrdU], which are added to cells (or animals) and then the incorporation of these precursors into genomic DNA during the S phase of the cell cycle (replication) is detected (Ruefli-Brasse et al., Science 302(5650):1581-4, 2003; Gu et al., Science 302 (5644):445-9, 2003).
Candidate compounds that reduce the survival of a neoplastic cell are also useful as anti-neoplasm therapeutics. Assays for measuring cell viability are known in the art, and are described, for example, by Crouch et al. (J. Immunol. Meth. 160, 81-8); Kangas et al. (Med. Biol.62, 338-43, 1984); Lundin et al., (Meth. Enzymol.133, 27-42, 1986); Petty et al.
(Comparison of J. Biolum. Chemilum.lO, 29-34, 1995); and Cree et al. (Anticancer Drugs 6: 398-404, 1995). Cell viability can be assayed using a variety of methods, including MTT (3- (4,5-dimethylthiazolyl)-2,5-diphenyltetrazolium bromide) (Barltrop, Bioorg. & Med. Chem. Lett.1: 611, 1991; Cory et al., Cancer Comm. 3, 207-12, 1991; Paull J. Heterocyclic Chem. 25, 911, 1988). Assays for cell viability are also available commercially. These assays include CELLTITER-GLO® Luminescent Cell Viability Assay (Promega), which uses luciferase technology to detect ATP and quantify the health or number of cells in culture, and the CellTiter-Glo® Luminescent Cell Viability Assay, which is a lactate dehyrodgenase (LDH) cytotoxicity assay. Candidate compounds that increase neoplastic cell death (e.g., increase apoptosis) are also useful as anti-neoplasm therapeutics. Assays for measuring cell apoptosis are known to the skilled artisan. Apoptotic cells are characterized by characteristic morphological changes, including chromatin condensation, cell shrinkage and membrane blebbing, which can be clearly observed using light microscopy. The biochemical features of apoptosis include DNA fragmentation, protein cleavage at specific locations, increased mitochondrial membrane permeability, and the appearance of phosphatidylserine on the cell membrane surface. Assays for apoptosis are known in the art. Exemplary assays include TUNEL (Terminal deoxynucleotidyl Transferase Biotin-dUTP Nick End Labeling) assays, caspase activity (specifically caspase-3) assays, and assays for fas-ligand and annexin V. Commercially
available products for detecting apoptosis include, for example, Apo-ONE® Homogeneous Caspase-3/7 Assay, FragEL TUNEL kit (ONCOGENE RESEARCH PRODUCTS, San Diego, CA)5 the ApoBrdU DNA Fragmentation Assay (BIOVISION, Mountain View, CA), and the Quick Apoptotic DNA Ladder Detection Kit (BIOVTSION, Mountain View, CA).
Microarrays
The methods of the invention may also be used for microarray-based assays that provide for the high-throughput analysis of cancer biomarkers. The ASPM nucleic acid molecules or polypeptides of the invention are useful as hybridizable array elements in such a microarray. The array elements are organized in an ordered fashion such that each element is present at a specified location on the substrate. Useful substrate materials include membranes, composed of paper, nylon or other materials, filters, chips, glass slides, and other solid supports. The ordered arrangement of the array elements allows hybridization patterns and intensities to be interpreted as expression levels of particular genes or proteins. Methods for making nucleic acid microarrays are known to the skilled artisan and are described, for example, in U.S. Pat. No. 5,837,832, Lockhart, et al. (Nat. Biotech. 14:1675-1680, 1996), and Schena, et al. (Proc. Natl. Acad. Sci. 93:10614-10619, 1996), herein incorporated by reference. Methods for making polypeptide microarrays are described, for example, by Ge (Nucleic Acids Res. 28:e3.i-e3.vii, 2000), MacBeath et al., (Science 289:1760-1763, 2000), Zhu et al. (Nature Genet. 26:283-289), and in U.S. Pat. No. 6,436,665, hereby incorporated by reference.
Nucleic Acid Microarrays
To produce a nucleic acid microarray oligonucleotides may be synthesized or bound to the surface of a substrate using a chemical coupling procedure and an ink jet application apparatus, as described in PCT application WO95/251116 (Baldeschweiler et al.), incorporated herein by reference. Alternatively, a gridded array may be used to arrange and link cDNA fragments or oligonucleotides to the surface of a substrate using a vacuum system, thermal, UV, mechanical or chemical bonding procedure.
A nucleic acid molecule (e.g. RNA or DNA) derived from a biological sample may be used to produce a hybridization probe as described herein. The biological samples are generally derived from a patient, preferably as a bodily fluid (such as blood, cerebrospinal fluid, phlegm, saliva, or urine) or tissue sample (e.g. a tissue sample obtained by biopsy). For some applications, cultured cells or other tissue preparations may be used. The mRNA is isolated according to standard methods, and cDNA is produced and used as a template to
make complementary RNA suitable for hybridization. Such methods are described herein. The RNA is amplified in the presence of fluorescent nucleotides, and the labeled probes are then incubated with the microarray to allow the probe sequence to hybridize to complementary oligonucleotides (e.g., ASPM nucleic acid molecules) bound to the microarray.
Incubation conditions are adjusted such that hybridization occurs with precise complementary matches or with various degrees of less complementarity depending on the degree of stringency employed. For example, stringent salt concentration will ordinarily be less than about 750 mM NaCl and 75 mM trisodium citrate, preferably less than about 500 mM NaCl and 50 mM trisodium citrate, and most preferably less than about 250 mM NaCl and 25 mM trisodium citrate. Low stringency hybridization can be obtained in the absence of organic solvent, e.g., formamide, while high stringency hybridization can be obtained in the presence of at least about 35% formamide, and most preferably at least about 50% formamide. Stringent temperature conditions will ordinarily include temperatures of at least about 300C, more preferably of at least about 37°C, and most preferably of at least about 42°C. Varying additional parameters, such as hybridization time, the concentration of detergent, e.g., sodium dodecyl sulfate (SDS), and the inclusion or exclusion of carrier DNA, are well known to those skilled in the art. Various levels of stringency are accomplished by combining these various conditions as needed. In one embodiment, hybridization will occur at 3O0C in 750 mM NaCl, 75 mM trisodium citrate, and 1% SDS. In another embodiment, hybridization will occur at 37°C in 500 mM NaCl, 50 mM trisodium citrate, 1% SDS, 35% formamide, and 100 μg/ml denatured salmon sperm DNA (ssDNA). In yet another embodiment, hybridization will occur at 42°C in 250 mM NaCl, 25 mM trisodium citrate, 1% SDS, 50% formamide, and 200 μg/ml ssDNA. Useful variations on these conditions will be readily apparent to those skilled in the art.
The removal of nonhybridized probes may be accomplished, for example, by washing. The washing steps that follow hybridization can also vary in stringency. Wash stringency conditions can be defined by salt concentration and by temperature. As above, wash stringency can be increased by decreasing salt concentration or by increasing temperature. For example, stringent salt concentration for the wash steps will preferably be less than about 30 mM NaCl and 3 mM trisodium citrate, and most preferably less than about 15 mM NaCl and 1.5 mM trisodium citrate. Stringent temperature conditions for the wash • steps will ordinarily include a temperature of at least about 25°C, at least about 42°C, or at least about 680C. In one embodiment, wash steps will occur at 25°C in 30 mM NaCl, 3 mM
trisodium citrate, and 0.1% SDS. In another embodiment, wash steps will occur at 42°C in 15 mM NaCl, 1.5 mM trisodium citrate, and 0.1% SDS. In yet another embodiment, wash steps will occur at 68°C in 15 mMNaCl, 1.5 mM trisodium citrate, and 0.1% SDS. Additional variations on these conditions will be readily apparent to those skilled in the art. A detection system may be used to measure the absence, presence, and amount of hybridization for all of the distinct sequences simultaneously (e.g., Heller et al., Proc. Natl. Acad. Sci. 94:2150-2155, 1997). Preferably, a scanner is used to determine the levels and patterns of fluorescence.
Protein Microarrays ASPM polypeptides (e.g., ASPM, ASPM variant 1, 2, or 3), such as those described herein, may also be analyzed using protein microarrays. Such arrays are useful in high- throughput low-cost screens to identify peptide or candidate compounds that bind a polypeptide of the invention, or fragment thereof. Typically, protein microarrays feature a protein, or fragment thereof, bound to a solid support. Suitable solid supports include membranes (e.g., membranes composed of nitrocellulose, paper, or other material), polymer- based films (e.g., polystyrene), beads, or glass slides. For some applications, ASPM polypeptides (e.g., ASPM, ASPM variant 1, 2, or 3) are spotted on a substrate using any convenient method known to the skilled artisan (e.g., by hand or by inkjet printer). Preferably, such methods retain the biological activity or function of the protein bound to the substrate (e.g., ASPM antibody binding).
The protein microarray is hybridized with a detectable probe. Such probes can be polypeptide (e.g., an ASPM antibody), nucleic acid, or small molecules. For some applications, polypeptide and nucleic acid probes are derived from a biological sample taken from a patient, such as a bodily fluid (such as blood, urine, saliva, or phlegm); a homogenized tissue sample (e.g. a tissue sample obtained by biopsy); or cultured cells (e.g., lymphocytes). Probes can also include antibodies, candidate peptides, nucleic acids, or small molecule compounds derived from a peptide, nucleic acid, or chemical library. Hybridization conditions (e.g., temperature, pH, protein concentration, and ionic strength) are optimized to promote specific interactions. Such conditions are known to the skilled artisan and are described, for example, in Harlow, E. and Lane, D., Using Antibodies: A Laboratory Manual. 1998, New York: Cold Spring Harbor Laboratories. After removal of non-specific probes, specifically bound probes are detected, for example, by fluorescence, enzyme activity (e.g., an enzyme-linked calorimetric assay), direct immunoassay, radiometric assay, or any other suitable detectable method known to the skilled artisan.
Detection of an increase in the amount of an ASPM polypeptide (e.g., ASPM, ASPM variant 1, 2, or 3) or an ASPM polynucleotide present in a patient sample is useful as a diagnostic for the presence of a neoplasia. Optionally, ASPM detection may be combined with the detection of other biomarkers, where the presence or level of the biomarker is correlated with the presence of a neoplasia.
Pharmaceutical Compositions
The present invention contemplates pharmaceutical preparations comprising an ASPM protein, a polynucleotide that encodes an ASPM protein, or an ASPM inhibitory nucleic acid molecule (e.g., a polynucleotide that hybridizes to and interferes with the expression of an ASPM polynucleotide), together with a pharmaceutically acceptable carrier. Polynucleotides of the invention may be administered as part of a pharmaceutical composition. The compositions should be sterile and contain a therapeutically effective amount of the polypeptides or nucleic acid molecules in a unit of weight or volume suitable for administration to a subject.
These compositions ordinarily will be stored in unit or multi-dose containers, for example, sealed ampoules or vials, as an aqueous solution or as a lyophilized formulation for reconstitution. As an example of a lyophilized formulation, 10 mL vials are filled with 5 mL of sterile-filtered 1% (w/v) aqueous ASPM polynucleotide solution, such as an aqueous solution of ASPM polynucleotide or polypeptide, and the resulting mixture can then be lyophilized. The infusion solution can be prepared by reconstituting the lyophilized material using sterile Water-for-Injection (WFI).
The ASPM polynucleotide, or polypeptide, or analogs may be combined, optionally, with a pharmaceutically acceptable excipient. The term "pharmaceutically-acceptable excipient" as used herein means one or more compatible solid or liquid filler, diluents or encapsulating substances that are suitable for administration into a human. The term "carrier" denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate administration. The components of the pharmaceutical compositions also are capable of being co-mingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction that would substantially impair the desired pharmaceutical efficacy.
The compositions can be administered in effective amounts. The effective amount will depend upon the mode of administration, the particular condition being treated and the desired outcome. It may also depend upon the stage of the condition, the age and physical
condition of the subject, the nature of concurrent therapy, if any, and like factors well known to the medical practitioner. For therapeutic applications, it is that amount sufficient to achieve a medically desirable result.
With respect to a subject having an neoplastic disease or disorder, an effective amount is sufficient to stabilize, slow, or reduce the proliferation of the neoplasm. Generally, doses of active polynucleotide compositions of the present invention would be from about 0.01 mg/kg per day to about 1000 mg/kg per day. It is expected that doses ranging from about 50 to about 2000 mg/kg will be suitable. Lower doses will result from certain forms of administration, such as intravenous administration. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits. Multiple doses per day are contemplated to achieve appropriate systemic levels of the ASPM polynucleotide or polypeptide compositions of the present invention.
A variety of administration routes are available. The methods of the invention, generally speaking, may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects. Other modes of administration include oral, rectal, topical, intraocular, buccal, intravaginal, intracisternal, intracerebroventricular, intratracheal, nasal, transdermal, within/on implants, e.g., fibers such as collagen, osmotic pumps, or grafts comprising appropriately transformed cells, etc., or parenteral routes. A particular method of administration involves coating, embedding or derivatizing fibers, such as collagen fibers, protein polymers, etc. with therapeutic proteins. Other useful approaches are described in Otto, D. et al., J. Neurosci. Res. 22: 83-91 and in Otto, D. and Unsicker, K. J. Neurosci. 10: 1912-1921.
Combination Therapies for the Treatment of a Neoplasm
Compositions and methods of the invention may be used in combination with any conventional therapy known in the art. In one embodiment, an ASPM polynucleotide or polypeptide composition of the invention having anti-neoplastic activity may be used in combination with any anti-neoplastic therapy known in the art. Exemplary anti-neoplastic therapies include, for example, chemotherapy, cryotherapy, hormone therapy, radiotherapy, and surgery. A ASPM polynucleotide composition of the invention may, if desired, include one or more chemotherapeutics typically used in the treatment of a neoplasm, such as abiraterone acetate, altretamine, anhydrovinblastine, auristatin, bexarotene, bicalutamide,
BMSl 84476, 2,3,4,5,6-pentafluoro-N-(3-fluoro-4-methoxyphenyl)benzene sulfonamide, bleomycin, N,N-dimethyl-L-valyl-L-valyl-N-methyl-L-valyl-L-proly- l-Lproline-t- butylamide, cachectin, cemadotin, chlorambucil, cyclophosphamide, 3',4'-didehydro-4'- deoxy-δ'-norvin- caleukoblastine, docetaxol, doxetaxel, cyclophosphamide, carboplatin, carmustine (BCNU),cisplatin, cryptophycin, cyclophosphamide, cytarabine, dacarbazine (DTIC), dactinomycin, daunorubicin, dolastatin, doxorubicin (adriamycin), etoposide, 5- fluorouracil, finasteride, flutamide, hydroxyurea and hydroxyureataxanes, ifosfamide, liarozole, lonidamine, lomustine (CCNU), mechlorethamine (nitrogen mustard), melphalan, mivobulin isethionate, rhizoxin, sertenef, streptozocin, mitomycin, methotrexate, 5- fluorouracil, nilutamide, onapristone, paclitaxel, prednimustine, procarbazine, RPR109881, stramustine phosphate, tamoxifen, tasonermin, taxol, tretinoin, vinblastine, vincristine, vindesine sulfate, and vinflunine. Other examples of chemotherapeutic agents can be found in Cancer Principles and Practice of Oncology by V. T. Devita and S. Hellman (editors), 6th edition (Feb. 15, 2001), Lippincott Williams & Wilkins Publishers.
The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the assay, screening, and therapeutic methods of the invention, and are not intended to limit the scope of what the inventors regard as their invention.
ASPM and Microcephaly
Primary autosomal recessive microcephaly is a genetic disorder in which an affected individual has a head circumference >3 standard deviations below the age- and sex-related mean and is mentally retarded, but typically has no other abnormal findings. Brain scans show that while the entire brain is reduced in size, the cerebral cortex is most severely affected. Because the vast majority of neurons are generated by week 21 of fetal life, microcephaly is likely due to a reduced production of neurons. The most common cause of microcephaly appears to be mutations in the ASPM gene.
The ASPM gene, which spans 65 kb of genomic DNA at Iq31, encodes a 10,434 base pair coding sequence (GenBank Accession No. NM_018136) containing 28 exons.
Expression studies in mice show ϊhatAspm is specifically expressed in the cerebral cortical •ventricular zone, the proliferative region of the lateral and medial ganglionic eminence, and the ventricular zone of the dorsal diencephalon at embryonic day 14.5. Expression diminishes by E16.5 and is greatly reduced at birth. Expression after birth was limited to
regions of the brain that showed persistent postnatal neurogenesis. Thus, the expression pattern oϊAspm suggests that it has an important role in regulating neurogenesis before and after birth.
The predicted ASPM protein has two distinguished regions, a tandem pair of N- terminal putative calponin-homology (CH) domains and a large block of IQ motifs (the single letter codes for the amino acids isoleucine (I) and glutamine (Q)), a sequence which mediates interactions with calmodulin and calmodulin-related proteins. CH domains are about 100 residues long and are commonly involved in actin binding, but many other substrates are found in vivo (Gimona et al., FEBS Lett. 513, 98-106, 2002; Korenbaum et al., J. Cell Sci. 115, 3543-3545, 2002). The IQ calmodulin-binding motif comprises 20-25 amino acids, with the core fitting the consensus IQXXXRGXXXR (where X is any amino acid) (Mooseker et al., Annual. Rev. Cell. Dev. Biol. 11, 633-675, 1995; Houdusse et al., Structure 4, 1475-1490, 1996). CH and IQ domains were first discovered in motor proteins such as unconventional myosins (Matin et al., Protein Sci. 12, 2909-2923). Calmodulin binding to IQ-motifs induces a conformational change in the proteins that regulates the binding of actin to the amino- terminal CH domains (Bahler et al., FEBS Lett. 513, 107-113). Typically unconventional myosins contain between 2 and 6 IQ motifs. They are not identical within a single protein, i.e. some of them have a higher affinity for the Ca+2 -free form of calmodulin, while other motifs have a higher affinity for the Ca+2/calmodulin. The number of IQ motifs and their divergence seems to determine the length of the lever arm and hence the step-size of myosin motor.
In contrast to unconventional myosins, ASPM does not have the catalytic Sl motor domain and therefore it cannot function as a mechanochemical protein. ASPM also differs from all known motor proteins by an extreme abundance of IQ motifs. The protein contains 81 putative calmodulin-binding motifs, most of which are encoded by exon 18, the largest exon in ASPM as reported herein.
The predicted ASPM protein is conserved between mammals, Drosophila, and worm, with a consistent correlation between nervous system complexity and protein length, principally involving an increase in the number of encoded IQ domains. For example, the Caenorhabditis elegans ASP homologue of ASPM contains 2 IQ domains; the Drosophila melonagaster ASP protein contains 24 repeats, and there are between 60-81 IQ domains in mammals. An interspecies sequence comparison reveals that the most highly diverged regions between primates and non-primate mammals are concentrated within the IQ array, suggesting that selection of specific segments of the ASPM gene may play a significant role
in brain evolution of mammals (Zhang et al., Genetics 165, 2063-2070, 2003; Evans et al., Hum. MoI. Genet. 13, 489-494, 2004; Kouprina et al., PIoS Biol. 5, E126, 2004).
Drosophila abnormal spindle (αspj-deficient mutants exhibit a mitotic metaphase checkpoint arrest with abnormal spindle poles, suggesting that Asp is required for the integrity of microtubule organizing centers. The Drosophila asp gene encodes a 220 kDa microtubule-associated protein found at the spindle poles and centrosomes from prophase to early telophase. Besides calmodulin-IQ-binding and calponin homology domains, the Asp protein has consensus phosphorylation sites for CDKl and MAP kinases (Saunders et al., J. Cell. Biol. 137, 881-890, 1997). Asp is co-purified with γ — tubulin from centrosomes and they are both required for the organization of microtubules into asters (Avides et al., Science 283, 1733-1735, 1999). This activity is dependent on the phosphorylation of Asp by the kinase Polo (Avides et al., Nat. Cell. Biol. 4, 421-424, 2001; Glover et al., Oncogene 24, 230-237, 2005). Together these observations suggest that mutations in human ASPM may cause microcephaly due to the disregulation of mitotic spindle activity in neuronal progenitor cells.
As reported in the Examples below, present studies now indicate that ASPM and alternatively spliced isoforms of ASPM are expressed in a variety of embryonic and adult tissues and are upregulated in neoplasia.
Example 1: ASPM is expressed in embryonic and adult tissues
Using in situ hybridization, previous studies detected expression of mouse ASPM in the cerebral cortical ventricular zone, the proliferative region of the lateral and medial ganglionic eminence and the ventricular zone o the dorsal diencephalon (Bond et al., Nat. Genet. 32, 316-320, 2005; Luers et al., Meek Dev. 118, 229-232, 2002). The present study investigated gene expression in the whole mouse embryo and found that Aspm is also expressed in during the development of a variety of organs, including the liver, heart, lung, and kidney (Figure IA). Similar results were obtained for human ASPM using RT-PCR analysis. Human ASPM transcripts were detected in a variety of different human embryonic tissues, including brain, bladder, colon, heart, liver, lung, skeletal muscle, skin, spleen, and stomach. Human expression was analyzed using two pairs of primers specific to exons 2 and 3, and exons 13 and 14 (FN2-3/RN2-3 and FN14-15/RN14-15, respectively). The nucleotide sequences of primers used in this work are provided at Table 1 (below) (SEQ ID Nos:98- 117).
Table !: Primers
Oligonucleotide name Sequence Size of RT-PCR product(s)
RT-PCR analysis of human ASPM transcripts Exon2-exon3
FN2-3/RN2-3 5'- ctgttaactggacaccactc -3V51- 258 bp tgtagtgggctcctaactct -3'
Exonl4-exonl5
FN14-15/RN14-15 5'-agacggccgtgtgttatgtt-3'/5'- 280 bp agcaggtattccaccaaggt -51
Exonl-exon28
ASPM1F/ASPM28R S'-tcctgtctctcagccacttc-S'/S1- 10,315 bp or 5,560 bp* atgccaagcgtatccatcac-3'
F2-3/33-18R1 S'-ctgttaactggacaccactc-S'/S1- 6,624 bp or 582 bp ctgcattttgtacctgaagga-3 '
F2-3/33-18R2 5'-ctgttaactggacaccactc-375'- 6,686 bp or 644 bp gtgcatctctcgcatccttt-3 '
Exl8R/Exl8F 5'-ataagcacgccaatgcctct-3'/5'- 4,355 bp ccttcagatggctgtgtatc-3 '
Exl8R/AS17F 5'-ataagcacgccaatgcctct-3'/5 '- 277 bp gagttaatgcagcactcgtc-3 '
Exl8F/AS19R S'-ccttcagatggctgtgtatc-S'/S '- 412 bp gcaggaagtatagctctcca-3 '
RT-PCR analysis of mouse Aspm transcripts
ASmIF/ ASm28R 5'-tgctgttgctcagccactt-3V5'- 9,217 bp or 5,574 bp cttccacattatcacgcctc-3'
ASmI 7F/ ASmI 9R 5'-aaggaagttgcggatgctga-3V5'- 4,095 bp or 300 bp acagaaagtgtagctctccatt-3 '
* The second product corresponds to an alternatively spliced variant. 258-bp and 239-bp bands of the expected sizes were detected in all samples, indicating that ASPM is ubiquitously expressed during development (Figure IB).
Because the Drosophila asp homologue is expressed in some adult tissues, human ASPM expression was analyzed in a variety of adult tissues, including breast, lung, pancreas, uterus, colon, thyroid, liver, bladder, kidney, ovary, testis, stomach, lymph node cervix, esophagus, and brain. Human ASPM transcripts were detected in all tissues except adult brain, though the level of expression was much lower than was observed in fetal tissues. The
ASPM expression analysis suggested that ASPM functions in non-CNS (non-central nervous system) tissues.
Example 2: ASPM expression is upregulated in cancer Expression data was obtained from over 120 uterine cancers and gene expression was analyzed using high density oligonucleotide microarrays. This analysis identified ASPM as one of several genes highly overexpressed in uterine cancer as compared to normal endometrium. Based on this initial observation, ASPM expression in normal and cancer tissues was analyzed by quantitative real-time RT-PCR. These studies confirmed that ASPM is more highly expressed in cancer of the uterus and ovary as compared to their expression in corresponding normal tissues (Figures 2A and 2B). The analysis of ASPM expression in cancer was expanded by including several other tumor types and obtained similar results for normal and matching tumor tissues of breast, colon, thyroid, testis, lymph, and stomach, as well as for 60 other cancer cell lines. These results are provided in Table 2, below, and in Figure 3 A.
Table 2 Expression of the ASPM gene in NIH-60 cancer cell lines
Sample number Type number Cancer type Cell line Expression
1 Leukemia K562 + 1 Leukemia MOLT4 + 1 Leukemia CCTF CEM + 1 Leukemia RPMI 8226 + 1 Leukemia HL-60 TB + 1 Leukemia SR + 2 CNS SF 268 + 2 CNS SF 295 +
9 2 CNS SF 539 +
10 2 CNS SNB 19 +
11 2 CNS SNB 75 +
12 2 CNS U251 +
13 3 Breast BT549 +
14 3 Breast HS 578 +
15 3 Breast MCF 7 +
16 3 Breast NCIADRRES +
18 3 Breast MDA MB 435 +
19 3 Breast T-47 D +
20 4 Colon Colo 205 +
21 4 Colon HCC 2998 +
22 4 Colon HCT 116 +
23 4 Colon HCT 15 +
24 4 Colon HT29 +
25 4 Colon KM12 +
26 4 Colon SW 620 +
27 5 NSCLC A549 ATCC +
28 5 NSCLC EKVX +
29 5 NSCLC HOP 62 +
30 5 NSCLC HOP 92 +
31 5 NSCLC NCI H 322 M +
32 5 NSCLC NCIH226 +
33 5 NSCLC NCI H23 +
34 5 NSCLC NCIH460 +
35 5 NSCLC NCIH522 +
36 6 Melanoma LOX IMVI +
37 6 Melanoma M14 +
38 6 Melanoma MAL ME +
54 6 Melanoma SK MEL 2 +
55 6 Melanoma SK MEL 5 +
56 6 Melanoma SK MEL 28 +
57 6 Melanoma UACC 62 +
58 6 Melanoma UACC 257 +
39 7 Ovarian IGROV 1 +
40 7 Ovarian OVCAR3 +
41 7 Ovarian OVCAR4 +
42 7 Ovarian OVCAR5 +
43 7 Ovarian OVCAR8 +
44 7 Ovarian SKOV3 +
45 Prostate DU 145 +
46 Prostate PC-3 +
47 Renal 786-0 +
48 Renal A498 +
49 Renal ACHN +
50 Renal CAKI-I +
51 Renal SN12C +
52 Renal TK-10 +
53 Renal UO-31 +
ASPM expression was checked by RT-PCR using primers FN2-3 and RN2-3 developed from exon 2 and exon 3 sequences (see Table 1).
Thus, transcription of the human ASPM gene is not restricted to developing brain. The ASPM gene is widely expressed in a variety of adult and embryo tissues and is upregulated in malignancy. Without being tied to one particular theory, it is possible that ASPM is upregulated in malignancy, because its expression correlates with the number of dividing cells in the malignant tissue. Alternatively, ASPM may be required in tumors that are increasing their size just as it is required for increases in brain size in microcephalic individuals.
Example 3: Alternatively spliced variants of human ASPM encode different numbers of IQ domains
The open reading frame (ORF) of ASPM was predicted based on the sequences of multiple small size Expressed Sequence Tags (ESTs) deposited into GenBank, which cover 28 exons. To characterize the human transcript(s), RT-PCR analysis was carried out using primers, ASPMlF and ASPM28R, which are present in the presumed first and last exons of ASPM (Table 1). As seen in Figured 4A and 4B (actin), two predominant PCR products were detected in RNA from fetal tissues. The largest of these RNAs migrated at a size consistent with the predicted full-length mRNA of 10,434 base pairs and was present in all fetal human tissues analyzed. The next most abundant band migrated at approximately 5.6 kb. Additional smaller PCR products were also observed. Similar sized bands were detected in normal as well as in matching tumor tissues (Figure 3A). Expression of mouse ASPM in normal murine tissues is shown in Figure 3B. The predominant PCR products were directly sequenced. The largest human PCR product corresponded to the predicted full-length ORF of 10,434 nucleotides. Further cloning and sequencing of the smaller RTPCR fragments revealed three human mRNAs with open reading frames (ORFs) of 5,678 bp, 4,259 bp and 3,189 bp (GenBank Accession numbers AY971956, AY971957 and AY971955) (Table 3, and Figures 8C-8H). Wild-type ASPM polypeptide and polynucleotide sequences are shown in Figures 8 A and 8B.
Table 3: ASPMcDNA clones and deposited accessions
Identification Species Accessions*
Human splice variant 1 Homo sapiens AY971956
Human splice variant 2 Homo sapiens AY971957
Human splice variant 3 Homo sapiens AY971955
Mouse splice variant 1 Mus musculus AY971958
*GenBaiik accessions correspond to alternatively spliced variants identified in fetal brain and adult tissues. So, in addition to the predicted 3477 amino acid residues protein, the ASPM gene may encode at least three isoforms (Figure 5A and 5B) containing 1892 (variant 1 corresponding
to the abundant RT-PCR product), 1389 (variant 2) and 1062 (variant 3) amino acid residues (Figures 8C-8H). The sequence of the wild-type human ASPM polypeptide and the ASPM polynucleotide is provided at Figures 8A and 8B, respectively. The amino acid and nucleic acid sequence of the ASPM variants is provided at Figures 8C-8H. The main difference between these shorter isoforms is primarily in the number of IQ domains (Figures 5A). Specifically, variant 1 lacks exon 18 (carrying 61 IQ domains) leaving this isoform with 14 total IQ domains. Variants 2 and 3 utilize different splice junctions within exon 18 and truncate parts of the IQ array. As a result of this splicing, variant 2 has 41 complete plus one incomplete IQ domains and variant 3 has 27 complete and one incomplete IQ domain. In addition, both variants 2 and 3 lack exons 4-17, which encode two CH (calponin-homology) domains and variant 2 also lacks exon 27, and has a stop codon in the intronic region after exon 26.
To investigate whether alternatively spliced ASPM isoforms are conserved in evolution, RT-PCR analysis was performed using a multiple mouse tissue cDNA panel and a pair of primers that hybridize to the first and last exons of murine Aspm. At least 2 predominant isoforms were detected on ethidium bromide stained gels (Figure 3B). Sequencing of these prominent bands indicated that the largest one was the full-length Aspm of 9,217 bp (containing 67 IQ domains) and the other was an exon 18 deleted isoform of 5,574 bp (15 IQ domains) transcript (accession number AY971958). Both transcripts were abundant in testis and in embryo. The presence of identical spliced ASPM variants lacking exon 18 in human and mouse suggested that this variant encodes the isoform that is required for ASPM function.
Example 4: IQ motifs in the ASPM protein are organized into a higher order repeat structure
More than half of the human ASPM protein consists of repeated calmodulin-binding IQ domains. The IQ array, which spans amino acid positions 1273-3234, is formed by 81 distinct IQ motifs of variable length (Figures 6A-6C). In comparison, the IQ array present in mouse and rat exhibits one large deletion corresponding to a region between IQs 57-70 in human, and several smaller insertions and deletions. In total, 67 IQ repeats are found in mouse Aspm (Figure 6D). The numbers of IQ repeats are slightly higher than previously reported, 74 and 61 repeats for human and mouse, respectively (Bond et al., Nat. Genet. 32, 316-320, 2005; Bond et al., Am. J. Hum. Genet. 73, 1170-1177, 2003) and the difference can be attributed to the very sensitive hidden Markov profile search used in our analysis.
The IQ repeat region of ASPM was examined. This analysis showed that the length of individual IQ domains is variable ranging from 14 (IQ 78) to 38 (IQ 76) amino acids (aa). The distribution of variably sized IQ repeats is of random (Figure 6A). The central part of the array between IQ 4-54 displays a striking periodicity with long (27 aa) IQ repeats being dispersed among short (23 aa) IQ repeats. The only exceptions are IQ 43 with 26 instead of 27 aa, and IQs 7-8 with 22 instead of 23aa. The spacing between the long and short units is highly regular. The long IQ 6 repeat is followed by four short units, IQ 11 and IQ 15 by three units. The next 12 long units are in each case followed by two short units. This region forms a highly regular array of 63 aa long superunits, composed of one long 27 aa unit and two short 23 aa units. The N-terminal and C-terminal parts of the IQ repeat region are less regular and contain IQ units of variable lengths. The mouse Aspm protein displays the same periodicity 27-23-23 in the central region of the protein. The N and C terminals of the protein are less organized as is the case of human ASPM (Figure 6D).
The sequence conservation of the long (Figure 6B) and short units (Figure 6C) was also of interest. The data indicated that the consensus sequence is (I,l,v)QX2(Y,F,w)(k,r)aXlo(y,f)X3(k,r)X3(i,l,v)X for the long unit; (I,l,v)QsX(Y,F,w)(R)Xi5(i,l,v)X for the short unit. Strongly conserved amino acids are denoted by uppercase letters, less well conserved/minor residues are in lowercase, and "X" ' denotes nonconserved positions. Physico-chemical properties of individual amino acid positions are shown in Figures 6B and 6C.
Finally the pattern of amino acid replacements during primate evolution was analyzed (Figure 6A). Positions identified as conserved in previous analyses were, as expected, mostly conserved among the primates analyzed. The majority of changes in primate ASPM sequences occurred in nonconserved residues. Both conservative and nonconservative substitutions are found in all long, short, and unordered repeats. Similar findings were obtained when the analysis was applied to mouse and rat ASPM sequences (Figure 6D). Interestingly, 66-69 IQ repeats, which are deleted in mice and rats. A higher-order repeat structure of IQ domains in all mammals indicates that this structure is likely to have functional significance. It is worth noting that because the IQ consensus sequence includes two positively charged amino acids, the IQ-containing region that spans 2,000 amino acids is highly positively charged. Among 530 charged residues, 467 are arginine and lysine. This density of basic residues may be required for binding of calmodulin, and/or it may facilitate interactions between ASPM and acidic proteins.
Example 5: Identification of a novel ASNP repeat region within ASPM
Analysis of ASPM revealed two novel repeats in the N-terminal part of the protein. The repeats are 32 and 35 amino acids long and localized to positions 316-347 and 366-400, respectively (Figures 5A and 5B). These repeats are highly conserved near the termini, with the central part being less well conserved. Similar repeats were found in all mammalian ASPM homologues. A single repeat was identified in the chicken gene. The interspecies conservation is similar to intraspecies comparisons, i.e. the termini are highly similar, the N- terminus is almost identical, but the central part is variable. The identified repeats are referred to as ASNP (for ASPM N-proximal) repeats. No significant similarity was identified between this motif and other proteins, even when using very sensitive searches (Position-
Specific Iterated Blast (PsiBlast), which was described by Altschul, et al. (Nucleic Acids Res. 25:3389-3402, 1997) and Schaffer, et al. (Nucleic Acids Res. 29: 2994-3005, 2001); and profile hidden Markov model (HMMER) using statistical descriptions of a sequence family's consensus for database searching, as described at the Washington University of St. Louis HMMER website that is maintained by Sean Eddy). Yet, the conservation in the compared species suggests that strong selective pressure preserves the repeats, indicating their functional importance for ASPM function.
Example 6: ASPM is found at the spindle poles during mitosis in human culture cells The role of Drosophila Asp in nucleating microtubules at centrosomes is consistent with its localization at the spindle poles during mitotic periods (Saunders et al., J. Cell. Biol. 137, 881-890, 1997). Polyclonal antibodies against three epitopes of human ASPM were generated. The anti-ASPM antibodies for two different epitopes, VTRK and QSPE, were used for indirect immunostaining of human HT1080 cultured cells. The VTRK antibody showed strong and similar typical spindle pole staining pattern during prometaphase, metaphase and anaphase (Figures 7A-I). A characterization of the VTRK antibody is shown at Figure 7 J, 7K, and 7L. A similar staining pattern was observed with the QSPE antibody during mitotic periods. The results of the mitotic subcellular localization and the highly conserved structural homology between Drosophila Asp and human ASPM suggested that human ASPM may be involved in a conserved spindle function.
As described herein, human ASPM is a mitotic spindle protein that is likely a functional orthologue of the Drosophila asp protein Bond et al., Nat. Genet. 32, 316-320 (6). This conclusion is supported by a recent analysis of proteins of the purified human mitotic spindles Sauer et al., (2005) Proteome analysis of the human mitotic spindle. MoI. Cell.
Protβomics 4, 35-43 (23) which provides evidence that microcephaly arises from deficient neurogenic mitosis. Without being tied to one particular theory, it is possible that as a component of the mitotic spindle, ASPM controls the proliferative symmetry of progenitors required for the expansion of cerebral cortical size Chenn et al., Cereb. Cortex 13, 599-606 (24). Alternatively, the presence of a nonfunctional ASPM might reduce chromosome segregation fidelity, resulting in a high incidence of chromosome aneuploidy, and reducing the ability of fetal stem cells to produce neurons.
Bond and co-authors identified two additional MCPH genes, MCPH3 and MCPH6 Bond et al., Nat. Genet. 37, 353-355 (4). It is notable that both of these genes encode spindle pole proteins. These findings, when taken together with the present studies, indicate a key role for the centrosome in the regulation of neurons generated by neural precursor cells. Identification of molecular partners of human ASPM would elucidate the molecular mechanisms controlling neuron generation. Such molecular partners might include the kinase Polo, which phosphorylates Drosophila Asp and co.-purifies with γ-tubulin, which is present in centrosomes Avides et al., Science 283, 1733-1735, Avides et al., Nat. Cell. Biol. 4, 421- 424, Saunders et al., The Drosophila gene abnormal spindle encodes a novel microtubule- associated protein that associates with the polar regions of the mitotic spindle. J. Cell. Biol. 137, 881-890 (18, 19, 21).
As for many other brain-expressed genes Andreadis, Biochim. Biophys. Ada. 1739, 91-103 (25), ASPM encodes several alternatively spliced mRNAs. The main ASPM isoform is a 3477 amino acid residue protein that contains eighty-one IQ motifs. Most of these are organized into a Higher-Order trimer Repeat (HOR) containing two units each comprising 23 and 27 amino acid residues. The HOR structure of the IQ array is conserved in primates and mouse ASPM proteins, suggesting that such a structure is essential for a protein function. Interestingly, five IQ repeats of myosin V are also organized into a higher-order structure, but the IQ domain is much shorter and forms a 25-23-25-23-25 amino acid residues array. Myosin V is an unconventional myosin that transports cellular cargos such as vesicles, melanosomes, or mRNA on actin filaments. The IQ periodicity seems to be necessary for efficient interaction between myosin V IQ domains and actin half-repeats, which are 36-nm long Sakamoto et al., J. Boil. Chem. 278, 29201-29207 (26). Analogously, the ASPM IQ periodicity may provide the exact spacing required for interactions with polymeric periodical proteins such as actin.
The major alternatively spliced form contains an in frame deletion of the entire exon 18 sequence and encodes 1892 amino acid residues with the predicted protein harboring fourteen flanking IQ domains that do not exhibit any periodicity. Similarly, two predominant transcripts were also present in mouse. In addition, alternatively spliced variants lacking both CH domains and more than a half of mostly periodical IQ domains were also detected, suggesting that ASPM may encode several proteins with different functions.
The presence of a long array of putative calmodulin-binding IQ domains is a unique feature of ASPM Bahler et al., (2002) FEBS Lett. 513, 107-113 (13). It has the highest number of the domains as compared to more than hundred IQ-containing proteins identified so far. Even if some of them are not functional, potentially ASPM could bind more calmodulin molecules than any other known IQ-containing protein. Calmodulin appears to regulate a protein function by modulating its ability to bind to other targets. This can occur by 2 mechanisms: i) by altering the conformation of the protein as well as ii) by influencing its subcellular location. Both the copy number and HOR structure of the calmodulin-binding domains in ASPM could greatly affect both mechanisms. The presence of multiple domains (IQ, CH and ASNP identified in this work) within ASPM also suggest its additional function such as a scaffolding protein that assembles multiprotein complexes.
If, similar to the Drosophila orthologue, human ASPM is involved in nucleating microtubules at centrosomes Saunders et al., The Drosophila gene abnormal spindle encodes a novel microtubule-associated protein that associates with the polar regions of the mitotic spindle. J. Cell. Biol. 137, 881-890, Avides et al., Science 283, 1733-1735, Avides et al., Nat. Cell. Biol. 4, 421-424, (20, 18, 19), a specific role of IQ repeats may be accumulation of Ca+2/calmodulins at the central region of the protein. Potentially one molecule of ASPM protein can bind through calmodulin several hundred calcium ions. Release of the bind Ca+2 may signal microtubule polymerization.
The results reported herein indicate that ASPM is widely expressed in adult tissues and is upregulated in cancer cells. The role of ASPM during mitosis of non-CNS (non-central nervous system) cells or its transcription may simply reflect proliferation of a tissue. Without being tied to one particular theory, given the localization of the ASPM gene product(s) at the mitotic spindle it is likely that some mutations (or polymorphic variants) that do not compromise a specific function of ASPM in neural progenitors may compromise the fidelity of cell division and cause chromosome instability in Adult tissues. Intriguingly, defects of the centrosome has been found in numerous forms of cancers Wang et al., DNA Cell. Biol. 23, 475-489, Marumoto T. et al., Nat. Rev. Cancer. 5, 42-50 (27, 28). Therefore, further
studies of ASPM polymorphism and function of the identified isofoπns will clarify molecular mechanisms of microcephaly, and determine whether specific mutations in the human ASPM nucleic acid sequence indicate a predisposition to cancer.
The experiments reported herein were carried out using the following materials and methods.
Mouse embryo in situ hybridization
Non-radioactive in situ hybridization was performed using a digoxigenin (DIG)- labelled cRNA probe. The antisense probe was generated from a mouse EST clone (Genbank accession: AW558815) using standard methods, and frozen sections hybridized and visualized using methods described previously (Berger et al., Jour. Comp. Neurol. 433, 101- 114, 2001).
Analysis of fetal and adult tissues by RT-PCR Total RNAs from fetal (brain, bladder, colon, testis, liver, heart, liver, skeletal muscle, skin, spleen, stomach), adult (brain, breast, lung, pancreas, uterus, colon, thyroid, liver, bladder, kidney, ovary, testis, stomach, lymph node, cervix, esophagus) and matching cancer human tissues, mouse (brain, testis, liver, heart) tissues (Ambion) were used for screening ASPM expression with the primers described in Table 1. cDNA was made from 1 μg of total RNA using a commercially available kit for reverse transcriptase polymerase chain reaction (RT-PCR), SUPERSCRIPT lsτ STRAND SYSTEM kit (Invitrogen, Carlsbad, CA), and priming with oligo dT per their standard protocol. Human β-actin primers (BD Biosciences Clontech, Palo Alto, CA) were used as positive controls for both human and mouse RT-PCR. NCI-60 cancer cell lines were from the National Cancer Institute, NIH. RT-PCR was performed using 1 μl of cDNA in a 50 ul reaction volume. Standard reaction conditions were: 94°C 5m, (94°C 1 minute, 55°C 1 minute, 72°C 1 minute x 35 cycles), 72°C 7 minutes, 4°C hold. Sequencing of the RT-PCR products confirmed their identity to the ASPM transcripts.
Analysis of cancer tissues by real-time PCR RNA levels of ASPM were assessed in clinical specimens. The relative expression of
ASPM was assayed by quantitative PCR using a commercially available assay system, FAM- labeled TaqMan® Gene Expression Assays, from Applied Biosystems, (Foster City, CA). Detectably labeled β-actin, VIC-labeled β-actin (4326315E), was used as a reference.
Samples were run on the ABI Prism® 7700 Sequence Detection System according to manufacturer's suggested Protocols in separate tubes. The relative quantitation, using the comparative Cx method, was calculated for each sample.
Sequencing of ASPM spliced forms
The full-length ASPM transcripts (10,434 bp for human and 9,846 bp for mouse) were sequenced after cloning of RT-PCR products into a TA cloning vector (Invitrogen, Carlsbad, CA). The RT-PCR products corresponding to human ASPM alternatively spliced variants of 5,679 bp, 4,259 bp and 3,189 bp and mouse spliced variant of 5,574 bp were also sequenced. Sequence forward and reverse reactions were run on a PE- Applied Biosystem 3100
Automated Capillary DNA Sequencer with a set of the designed primers. Comparative analyses were performed with the wild-type cDNA sequences (Genbank accession number gi:24211028 and gi:36031058 for human and mouse) using a publically available sequence analysis software, GCG DNA Analysis Wisconsin Package and NCBI BLAST. All sequenced clones were named and numbered according to the clone/accession identifier (Table 3).
Sequence analysis and identification of IQ repeats
Sequences were aligned by Dialign2.1 Morgenstern, B. (1999) Bioinformatics 15, 211-218 (30); http://bibiserv.techfak.uni-bielefeld.de/dialign/. Protein alignments were visualized using publically available software, GeneDoc (Nicholas et al., EMBNEW.NEWS 4, 14); http://www.psc.edu/biomed/ genedoc and WAVis [(Zika et al., Nucl. Acids Res. 32:W48-49, 2004); http://wavis.img.cas.cz. Conserved domains in the ASPM protein were detected by Rpsblast with default parameters (Marchler-Bauer et al., Nucleic Acids Res. 32, W48-49); http://www.ncbi.nlm.nih.gov/Structure/cdd/wrpsb.cgi]. The human ASPM protein was initially screened for IQ repeats by Radar fhttp://www.ebi.ac.uk/Radar/). The detected IQ repeats were used to build a hidden Markov profile using publically available software, HMMER v 2.2 Eddy, Bioinformatics 14, 755-763 (34); http://hmmer.wustl.edu/. In the next step, the obtained profile was used to search for the remaining IQ repeats. The IQ sequences were manually edited in order to obtain exact boundaries of all IQ units. This complete set was used to build a new hidden Markov profile, which subsequently served for detection of IQ repeats in mouse Aspm. Other ASPM repeats, including newly detected N-terminal repeats were identified by Radar.
Peptide specific antibodies and Western blot analysis
Three synthetic peptides representing amino acids 422-441
(QSPEDWRKSEVSPRIPE), 497-520 (VTKRKATCTRENOTEINKPKAKR), and 3443- 3463 (SRLKPDWVLRRDNMEEITNPL) from the ASPM sequence were conjugated to Keyhole Limpet Hemocyanin and used as immunogenes as previously described (Goldsmith , et al., Biochemistry 27, 7085-7090, 1988). The resulting antisera were affinity purified over columns of peptide conjugated to Affigel 15 (Bio-Rad, Richmond, CA), and concentrated in stirred cells with YM30 membranes (Millipore, Billerica, MA). The concentrates were then subjected to gel filtration chromatography using 2.6 x 60 cm SUPERDEX 200 columns (GE Healthcare, Piscataway, NJ) and the monomeric IgG fractions were pooled and concentrated. The protein concentrations were then determined using a commercially available kit, the Bradford assay (Bio-Rad, Richmond, CA). These peptides were identical to both human and mouse ASPM proteins. The antibodies specificity for these peptides was first confirmed with recombinant ASPM fragments. Plasmids for ASPM expression in E.coli cells were constructed by insertion of a 1353 bp fragment containing QSPE and VTKR epitopes, and a 705 bp fragment containing SRLK epitope. The fragments were PCR amplified from the full-size ASPM cDNA (positions 346-1695 and 9730-10434) and cloned into BatήBI site of the pMAL-p2X expression vector (BioLabs Inc.). To analyze ASPM protein in mammalian cells, human (HTl 080) and mouse (3T3) cells were mixed with SDS sample buffer containing a protease inhibitor cocktail (Sigma), homogenized using a 27 gauge needle and resolved in a 3.4% of 29:1 acrylamide/bis-acrylamide gel. Following electrophoresis, the proteins were transferred to PVDF membranes (Millipore) for 40 minutes at 15V in transfer buffer (50 mM Tris, 380 mM glycine, 0.1% SDS and 20% methanol) using a semi-dry method. All subsequent steps were carried out in phosphate buffered saline (PBS) containing 0.05% Tweet' 20 (TPBS). After thirty minutes blocking with 10% TPBS nonfat milk, the membranes were exposed for 12 hours to 1/2,000 diluted anti-QSPE or SRLK antibodies. The PVDF membrane was washed three times with TPBS, then incubated with anti-rabbit IgG conjugated with horse radish peroxidase (HRP) diluted 1/5000 for 30 minutes, and then washed as in the previous step. The membranes were incubated for 1 minute with commercially available chemiluminescent detection reagents, ECL plus reagents (Amersham) and exposed to Hperfilm ECL (Amersham). Immunoblotting of cellular proteins from human HTl 080 and mouse 3T3 cells revealed the predicted protein isoforms corresponding to two main alternatively spliced variants. These bands were not detected in a lymphoblast cell line derived from a microcephalic individual with a frameshift mutation in ASPM (Bond et al.,
Am. J. Hum. Genet. 73, 1170-1177) indicating that the antibodies are highly specific (Figure 7J, K5 L).
Indirect immunofluorescence staining with anti-ASPM antibodies For detection of the endogenous ASPM protein, two different fixation methods were employed with HT1080 cells grown on poly-L-lysine coating coverslips. 1) Cultured cells were first incubated for 15 minutes with 2% paraformaldehyde. Samples were washed twice and then treated for 5 minutes with 0.5% Triton X-100 (Sigma) and for 5 minutes with 0. IM Glycine (Sigma). 2) Cultured cells were treated for 30 minutes with 100% methanol (Mallinckrodt Baker). Fixed samples were incubated for 1 hour at 37°C with anti-ASPM antibodies (1 :200). After washing three times with PBS for 5 minutes each, samples were incubated for 1 hour at 37°C with a commercially available fluorescent dye, Alexa Fluor 594 coupled to goat anti-rabbit IgG (Molecular Probes). After washing three times with PBS for 5 minutes each, samples were stained with lμg/ml of DAPI and rinsed again with PBS. Samples were finally mounted using Vectashield mounting medium (Vector Lab). Images were captured using a Zeiss microscope (Axiophoto) equipped with a cooled-CCD camera (Cool SNAP HQ, Photometries Ltd) and analyzed by IPLab software (Signal Analytics).
Other Embodiments A number of embodiments of the invention have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are within the scope of the following claims
The practice of the present invention employs, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are well within the purview of the skilled artisan. Such techniques are explained fully in the literature, such as, "Molecular Cloning: A Laboratory Manual", second edition (Sambrook, 1989); "Oligonucleotide Synthesis" (Gait, 1984); "Animal Cell Culture" (Freshney, 1987); "Methods in Enzymology" "Handbook of Experimental Immunology" (Weir, 1996); "Gene Transfer Vectors for Mammalian Cells" (Miller and Calos, 1987); "Current Protocols in Molecular Biology" (Ausubel, 1987); "PCR: The Polymerase Chain Reaction", (Mullis, 1994); "Current Protocols in Immunology" (Coligan, 1991). These techniques are applicable
to the production of the polynucleotides and polypeptides of the invention, and, as such, may be considered in making and practicing the invention.
Claims
1. A method of diagnosing a subject as having, or having a propensity to develop, a neoplasia, the method comprising determining the level of expression of an ASPM nucleic acid molecule in a subject sample, wherein an increased level of expression relative to a reference, indicates that the subject has or has a propensity to develop a neoplasia.
2. A method of diagnosing a subject as having, or having a propensity to develop, a neoplasia, the method comprising determining the level of expression of an ASPM polypeptide in a subject sample, wherein an increased level of expression relative to the level of expression in a reference, indicates that the subject has or has a propensity to develop a neoplasia.
3. The method of claim 2, wherein the method comprises determining the level of expression of the ASPM polypeptide.
4. The method of claim 3, wherein the level of expression is determined in an immunological assay.
5. A method of diagnosing a subject as having, or having a propensity to develop, a neoplasia, the method comprising determining the level of biological activity of an ASPM polypeptide in a subject sample, wherein an alteration in the level of biological activity relative to the biological activity in a reference, indicates that the subject has or has a propensity to develop a neoplasia.
6. A method of monitoring a subject diagnosed as having a neoplasia, the method comprising determining the level of activity of an ASPM polypeptide in a subject sample, wherein an alteration in the level of expression relative to the level of expression in a reference indicates the severity of neoplasia in the subject.
7. The method of claim 6, wherein the subject is being treated for a neoplasia.
8. The method of claim 7, wherein the neoplasia is breast, prostate, lung, testis, ovary, or uterine neoplasia.
9. The method of claim 6, wherein the alteration is an increase, and the increase indicates an increased severity of neoplasia.
10. The method of any one of claims 1-6, wherein the reference is a control subject sample.
11. The method of claim 6, wherein the reference is a subject sample obtained at an earlier time point.
12. The method of any one of claims 1-6, wherein the subject sample is a biological sample.
13. The method of any one of claims 1-7, wherein the method is used to diagnose a subject as having neoplasia.
14. The method of any one of claims 1-7, wherein the method is used to determine the treatment regimen for a subject having neoplasia.
15. The method of any one of claims 1-7, wherein the method is used to monitor the condition of a subject being treated for neoplasia.
16. The method of any one of claims 1 -7, wherein the method is used to determine the prognosis of a subject having neoplasia.
17. The method of claim 16, wherein a poor prognosis determines an aggressive treatment regimen for the subject.
18. A method for identifying a subject as having or having a propensity to develop a neoplasia, the method comprising detecting an alteration in the sequence of an ASPM nucleic acid molecule relative to the sequence or expression of a reference molecule.
19. The method of claim 18, wherein the alteration is detected using a hybridization reaction.
20. An ASPM antibody that specifically binds to an ASPM protein or fragment thereof.
21. The antibody of claim 20, wherein the antibody binds to a VTRK or QSPE epitope of an ASPM polypeptide.
22. A polypeptide comprising an isolated ASPM protein variant, or fragment thereof, having substantial identity to ASPM variant 1, 2, or 3, wherein the variant is upregulated in a neoplastic cell.
23. The polypeptide of claim 22, wherein the ASPM protein variant is at least 85% identical to ASPM variant 1, 2, or 3.
24. The polypeptide of claim 22, wherein the ASPM protein variant comprises at least an IQ domain and is capable of binding calmodulin.
25. The polypeptide of claim 22, wherein the fragment consists of an ASPM protein variant selected from the group consisting of ASPM variant 1, 2, and 3.
26. The polypeptide of any one of claims 22-25, wherein the polypeptide is a fusion protein.
27. The polypeptide of any one of claims 22-25, wherein the polypeptide is linked to a detectable amino acid sequence.
28. The polypeptide of any one of claims 22-25, wherein the polypeptide is linked to an affinity tag.
29. An isolated ASPM nucleic acid molecule, wherein the nucleic acid molecule encodes a polypeptide of any one of claims 22-28.
30. A vector comprising the nucleic acid molecule of claim 29.
31. An isolated ASPM inhibitory nucleic acid molecule, wherein the inhibitory nucleic acid molecule specifically binds at least a fragment of a nucleic acid molecule encoding an
ASPM protein.
32. A vector comprising a nucleic acid molecule encoding the nucleic acid molecule of claim 31.
32. The vector of claim 30 or 32, wherein the vector is an expression vector.
33. The vector of claim 30 or 32, wherein the nucleic acid molecule is positioned for expression.
34. The vector of claim 30 or 32, wherein the nucleic acid molecule is operably linked to a promoter.
35. The vector of claim 30 or 32, wherein the promoter is suitable for expression in a mammalian cell.
36. A host cell comprising a nucleic acid molecule of any one of claims 29-35.
37. The host cell of claim 36, wherein the cell expresses an ASPM protein variant.
38. The host cell of claim 36, wherein the cell is in vitro.
39. The host cell of claim 36, wherein the cell is in vivo.
40. The host cell of claim 36, wherein the cell is a mammalian cell.
41. The host cell of claim 36, wherein the cell is a human cell.
42. A double-stranded RNA corresponding to at least a portion of an ASPM nucleic acid molecule that encodes an ASPM protein, wherein the double-stranded RNA is capable of altering the level of protein encoded by the ASPM nucleic acid molecule.
43. The RNA of claim 42, wherein the RNA is an siRNA.
44. An antisense nucleic acid molecule, wherein the antisense nucleic acid molecule is complementary to at least six nucleotides of an ASPM nucleic acid molecule that encodes an ASPM protein, and wherein the antisense is capable of altering expression from the nucleic acid molecule to which it is complementary.
45. A primer capable of binding to an ASPM nucleic acid molecule encoding an ASPM protein variant that is upregulated in a neoplastic tissue.
46. A collection of primers capable of binding to and amplifying an ASPM nucleic acid molecule, wherein at least one of the primers in the collection is the primer of claim 22.
47. A pharmaceutical composition comprising an effective amount of an ASPM protein, variant, or fragment thereof, in a pharmaceutically acceptable excipient, wherein the fragment is capable of modulating cell proliferation or chromosome stability.
48. A pharmaceutical composition comprising an effective amount of a nucleic acid molecule of any one of claims 29-44 in a pharmaceutically acceptable excipient, wherein the fragment is capable of modulating cell proliferation or chromosomal stability.
49. A pharmaceutical composition comprising an effective amount of a vector comprising a nucleic acid molecule encoding an ASPM protein of any one of claims 29-44 in a
pharmaceutically acceptable excipient, wherein expression of the ASPM protein in the cell is capable of modulating cell proliferation or chromosomal stability.
50. The pharmaceutical composition of claim 49, wherein the ASPM nucleic acid molecule is positioned for expression in a mammalian cell.
51. An ASPM biomarker purified on a biochip.
52. A microarray comprising at least two nucleic acid molecules, or fragments thereof, fixed to a solid support, wherein at least one of the nucleic acid molecules is an ASPM nucleic acid molecule.
53. A microarray comprising at least two polypeptides, or fragments thereof, bound to a solid support, wherein at least one of the polypeptides on the support is an ASPM polypeptide.
54. A diagnostic kit for the diagnosis of a neoplasia in a subject comprising an ASPM nucleic acid molecule, or fragment thereof, and written instructions for use of the kit for detection of a neoplasia.
55. A diagnostic kit for the diagnosis of a neoplasia in a subject comprising an antibody that specifically binds an ASPM polypeptide, or fragment thereof, and written instructions for use of the kit for detection of a neoplasia.
56. A kit identifying a subject as having or having a propensity to develop a neoplasia, comprising an adsorbent, wherein the adsorbent retains an ASPM biomarker, and written instructions for use of the kit for detection of a neoplasia.
57. A kit comprising a first capture reagent that specifically binds an ASPM biomarker, and written instructions for use of the kit for detection of a neoplasia.
58. A method for detecting neoplasia in a subject sample, the method comprising
(a) contacting a subject sample with a capture reagent affixed to a substrate; and
(b) capturing an ASPM polypeptide or nucleic acid molecule with the capture reagent.
59. The method of claim 58, wherein the subject sample comprises an ASPM protein or fragment thereof.
60. The method of claim 58, wherein the proteins are fractionated prior to contacting the capture reagent.
61. A method of altering the expression of an ASPM nucleic acid molecule in a cell, the method comprising contacting the cell with an effective amount of a compound capable of altering the expression of the ASPM nucleic acid molecule .
62. The method of claim 61 , wherein the compound is an ASPM antisense nucleic acid molecule, a small interfering RNA (siRNA), or a double stranded RNA (dsRNA) that inhibits the expression of an ASPM nucleic acid molecule.
63. A method of altering ASPM protein expression in a cell, the method comprising contacting the cell with a compound capable of altering the expression of an ASPM polypeptide.
64. The method of claim 61 or 63, wherein the cell is a human cell.
65. The method of claim 61 or 63, wherein the cell is a neoplastic cell.
66. The method of claim 61 or 63, wherein the cell is in vitro.
61. The method of claim 61 or 63, wherein the cell is in vivo.
68. A method of treating or preventing a neoplasia, the method comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition that alters expression of an ASPM polypeptide.
69. A method of identifying a compound that inhibits a neoplasia, the method comprising contacting a cell that expresses an ASPM nucleic acid molecule with a candidate compound, and comparing the level of expression of the nucleic acid molecule in the cell contacted by the candidate compound with the level of expression in a control cell not contacted by the candidate compound, wherein an alteration in expression of the ASPM nucleic acid molecule identifies the candidate compound as a compound that inhibits a neoplasia.
70. The method of claim 69, wherein the alteration in expression is a decrease in transcription.
71. The method of claim 69, wherein the alteration in expression is a decrease in translation.
72. A method of identifying a compound that inhibits a neoplasia, the method comprising contacting a cell that expresses an ASPM polypeptide with a candidate compound, and comparing the level of expression of the polypeptide in the cell contacted by the candidate compound with the level of polypeptide expression in a control cell not contacted by the candidate compound, wherein an alteration in the expression of the ASPM polypeptide identifies the candidate compound as a compound that inhibits a neoplasia.
73. A method of identifying a compound that inhibits a neoplasia, the method comprising contacting a cell that expresses an ASPM polypeptide with a candidate compound, and comparing the biological activity of the polypeptide in the cell contacted by the candidate compound with the level of biological activity in a control cell not contacted by the candidate
compound, wherein an alteration in the biological activity of the ASPM polypeptide identifies the candidate compound as a candidate compound that inhibits a neoplasia.
74. The method of any one of claims 68-73, wherein the cell is in vitro.
75. The method of any one of claims 68-73, wherein the cell is in vivo.
76. The method of any one of claims 68-73, wherein the cell is a human cell.
77. The method of any one of claims 68-73, wherein the cell is a neoplastic cell.
78. The method of any one of claims 68-73, wherein the alteration in expression is assayed using an immunological assay, an enzymatic assay, or a radioimmunoassay.
79. A method of identifying a candidate compound that inhibits a neoplasia, the method comprising a) contacting a cell containing an ASPM nucleic acid molecule present in an expression vector that includes a reporter construct; b) detecting the level of reporter gene expression in the cell contacted with the candidate compound with a control cell not contacted with the candidate compound, wherein an alteration in the level of the reporter gene expression identifies the candidate compound as a candidate compound that inhibits a neoplasia.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US69621205P | 2005-07-01 | 2005-07-01 | |
US60/696,212 | 2005-07-01 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2007005635A2 true WO2007005635A2 (en) | 2007-01-11 |
WO2007005635A3 WO2007005635A3 (en) | 2007-08-02 |
Family
ID=37605051
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2006/025640 WO2007005635A2 (en) | 2005-07-01 | 2006-06-29 | Mitotic spindle protein aspm as a diagnostic marker for neoplasia and uses therefor |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2007005635A2 (en) |
Cited By (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20110229524A1 (en) * | 2010-03-19 | 2011-09-22 | Immatics Biotechnologies Gmbh | Composition of tumor-associated peptides and related anti-cancer vaccine for the treatment of Gastric Cancer and other cancers |
WO2011113819A3 (en) * | 2010-03-19 | 2011-11-10 | Immatics Biotechnologies Gmbh | Diagnosis and treatment of cancer based on avl9 |
US8609608B2 (en) | 2007-01-16 | 2013-12-17 | C3 Jian, Inc. | Antimicrobial peptides |
WO2014186773A1 (en) * | 2013-05-17 | 2014-11-20 | National Health Research Institutes | Methods of prognostically classifying and treating glandular cancers |
US20210261636A1 (en) * | 2020-02-14 | 2021-08-26 | Janssen Biotech, Inc. | Neoantigens expressed in multiple myeloma and their uses |
Family Cites Families (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP4643450B2 (en) * | 2003-08-08 | 2011-03-02 | 株式会社ペルセウスプロテオミクス | Cancer high expression gene |
-
2006
- 2006-06-29 WO PCT/US2006/025640 patent/WO2007005635A2/en active Application Filing
Non-Patent Citations (3)
Title |
---|
DATABASE Geneseq [Online] EBI; 5 May 2005 (2005-05-05), ABURATANI H ET AL.: "Novel protein encoded by any one of TEG1 to TEG64, useful for diagnosing and treating cancer e.g. lung, hepatic, stomach, colon or pancreatic cancer." XP002433858 retrieved from WWW.EBI.CO.UK Database accession no. ADX83135 & WO 2005/014818 A (ABURATANI HIROYUKI [JP]; PERSEUS PROTEOMICS INC [JP]; CHUGAI PHARMACEU) 17 February 2005 (2005-02-17) * |
DATABASE GENESEQ [Online] EBI; 5 May 2005 (2005-05-05), ABURATANI H ET AL.: "Novel protein encoded by any one of TEG1 to TEG64, useful for diagnosing and treating cancer e.g. lung, hepatic, stomach, colon or pancreatic cancer." XP002433859 retrieved from WWW.EBI.CO.UK Database accession no. ADX83191 & WO 2005/014818 A (ABURATANI HIROYUKI [JP]; PERSEUS PROTEOMICS INC [JP]; CHUGAI PHARMACEU) 17 February 2005 (2005-02-17) * |
KOUPRINA NATALAY ET AL: "The microcephaly ASPM gene is expressed in proliferating tissues and encodes for a mitotic spindle protein" HUMAN MOLECULAR GENETICS, vol. 14, no. 15, 22 June 2005 (2005-06-22), pages 1-34, XP002433851 ISSN: 0964-6906(print) 1460-2083(ele * |
Cited By (42)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US8609608B2 (en) | 2007-01-16 | 2013-12-17 | C3 Jian, Inc. | Antimicrobial peptides |
AU2008251748B2 (en) * | 2007-01-16 | 2014-02-27 | C3 Jian, Inc. | Novel antimicrobial peptides |
US11298404B2 (en) | 2010-03-19 | 2022-04-12 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US9993523B2 (en) | 2010-03-19 | 2018-06-12 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US12064465B2 (en) | 2010-03-19 | 2024-08-20 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
EP2845604A3 (en) * | 2010-03-19 | 2015-05-20 | immatics biotechnologies GmbH | Novel immunotherapy against several tumors including gastrointestinal and gastric cancer |
US9101585B2 (en) | 2010-03-19 | 2015-08-11 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US9132177B2 (en) * | 2010-03-19 | 2015-09-15 | Immatics Biotechnologies Gmbh | Composition of tumor-associated peptides and related anti-cancer vaccine for the treatment of gastric cancer and other cancers |
JP2016034950A (en) * | 2010-03-19 | 2016-03-17 | イマティクス バイオテクノロジーズ ゲーエムベーハー | Novel immunotherapy for several tumors including gastrointestinal cancer and gastric cancer |
US11975042B2 (en) | 2010-03-19 | 2024-05-07 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US11969455B2 (en) | 2010-03-19 | 2024-04-30 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US11957730B2 (en) | 2010-03-19 | 2024-04-16 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
EA024497B1 (en) * | 2010-03-19 | 2016-09-30 | Имматикс Байотекнолоджиз Гмбх | PEPTID, CONNECTING WITH THE MOLECULE OF THE MAIN COMPLEX OF THE HYDROCABILITY OF THE PERSON OF I CLASS, AND ITS APPLICATION FOR THE TREATMENT OF CANCER |
US11883462B2 (en) | 2010-03-19 | 2024-01-30 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US9717774B2 (en) | 2010-03-19 | 2017-08-01 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US9895415B2 (en) | 2010-03-19 | 2018-02-20 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US10478471B2 (en) | 2010-03-19 | 2019-11-19 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US10064913B2 (en) | 2010-03-19 | 2018-09-04 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US10159725B2 (en) | 2010-03-19 | 2018-12-25 | Immatics Biotechnologies Gmbh | Composition of tumor-associated peptides and related anti-cancer vaccine for the treatment of gastric cancer and other cancers |
US10357540B2 (en) | 2010-03-19 | 2019-07-23 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
WO2011113819A3 (en) * | 2010-03-19 | 2011-11-10 | Immatics Biotechnologies Gmbh | Diagnosis and treatment of cancer based on avl9 |
US10420816B1 (en) | 2010-03-19 | 2019-09-24 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US11077171B2 (en) | 2010-03-19 | 2021-08-03 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US10898546B2 (en) | 2010-03-19 | 2021-01-26 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US10905741B2 (en) | 2010-03-19 | 2021-02-02 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US10933118B2 (en) | 2010-03-19 | 2021-03-02 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
CN112430255A (en) * | 2010-03-19 | 2021-03-02 | 伊玛提克斯生物技术有限公司 | Novel immunotherapy for several tumors including gastrointestinal and gastric cancers |
TWI715332B (en) * | 2010-03-19 | 2021-01-01 | 德商艾瑪提克生物技術有限公司 | Novel immunotherapy against several tumors including gastrointestinal and gastric cancer |
US11850274B2 (en) | 2010-03-19 | 2023-12-26 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US11273200B2 (en) | 2010-03-19 | 2022-03-15 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US20110229524A1 (en) * | 2010-03-19 | 2011-09-22 | Immatics Biotechnologies Gmbh | Composition of tumor-associated peptides and related anti-cancer vaccine for the treatment of Gastric Cancer and other cancers |
TWI766362B (en) * | 2010-03-19 | 2022-06-01 | 德商艾瑪提克生物技術有限公司 | Novel immunotherapy against several tumors including gastrointestinal and gastric cancer |
TWI766361B (en) * | 2010-03-19 | 2022-06-01 | 德商艾瑪提克生物技術有限公司 | Novel immunotherapy against several tumors including gastrointestinal and gastric cancer |
TWI768461B (en) * | 2010-03-19 | 2022-06-21 | 德商艾瑪提克生物技術有限公司 | Novel immunotherapy against several tumors including gastrointestinal and gastric cancer |
US11648292B2 (en) | 2010-03-19 | 2023-05-16 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
US11839643B2 (en) | 2010-03-19 | 2023-12-12 | Immatics Biotechnologies Gmbh | Immunotherapy against several tumors including gastrointestinal and gastric cancer |
TWI560275B (en) * | 2013-05-17 | 2016-12-01 | Nat Health Research Institutes | Methods and kits of prognostically classifying and use of manufacturing pharmaceutical composition using nucleic acid for inhibiting cancer stem cell aggregation or tumor migration |
JP2016525883A (en) * | 2013-05-17 | 2016-09-01 | ナショナル ヘルス リサーチ インスティテュートス | Prognostic classification and treatment of adenocarcinoma |
CN105473772A (en) * | 2013-05-17 | 2016-04-06 | 财团法人国家卫生研究院 | Prognostic Classification and Approach to Glandular Cancer |
US20160090638A1 (en) * | 2013-05-17 | 2016-03-31 | National Health Research Institutes | Methods of prognostically classifying and treating glandular cancers |
WO2014186773A1 (en) * | 2013-05-17 | 2014-11-20 | National Health Research Institutes | Methods of prognostically classifying and treating glandular cancers |
US20210261636A1 (en) * | 2020-02-14 | 2021-08-26 | Janssen Biotech, Inc. | Neoantigens expressed in multiple myeloma and their uses |
Also Published As
Publication number | Publication date |
---|---|
WO2007005635A3 (en) | 2007-08-02 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US12084723B2 (en) | Compositions and methods for treating or preventing prostate cancer and for detecting androgen receptor variants | |
Kouprina et al. | The microcephaly ASPM gene is expressed in proliferating tissues and encodes for a mitotic spindle protein | |
US20070243176A1 (en) | Human genes and gene expression products | |
US20060228710A1 (en) | Novel therapeutic targets in cancer | |
WO2007005635A2 (en) | Mitotic spindle protein aspm as a diagnostic marker for neoplasia and uses therefor | |
EP1507790A2 (en) | Human genes and gene expression products isolated from human prostate | |
US20070212351A1 (en) | Novel therapeutic targets in cancer | |
US7078188B2 (en) | MUC17 encoding nucleic acid sequences, polypeptides, antibodies and methods of use thereof | |
EP1144636A2 (en) | Human genes and gene expression products | |
EP1379651A2 (en) | Human genes and gene expression products isolated from human prostate | |
US9540694B2 (en) | HEYL as a therapeutic target and a diagnostic marker for neoplasia and uses therefor | |
US7867978B2 (en) | Methods and compositions for treating neurological disorders | |
CN105331686B (en) | The preparation of Myosin9b protein specific antibodies and Myosin9b gene families change the application in diagnosing tumor and prognosis | |
AU2002354947B2 (en) | Methods for diagnosing and treating diseases and conditions of the digestive system and cancer | |
US6372467B1 (en) | P54s6k and p85s6k genes, proteins, primers, probes, and detection methods | |
AU2002354947A1 (en) | Methods for diagnosing and treating diseases and conditions of the digestive system and cancer | |
JP2006254830A (en) | Material for suppressing sept11 gene expression |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
NENP | Non-entry into the national phase in: |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 06785991 Country of ref document: EP Kind code of ref document: A2 |