WO2024133723A1 - Methods for decreasing therapeutic acquired resistance to chemotherapy and/or radiotherapy - Google Patents
Methods for decreasing therapeutic acquired resistance to chemotherapy and/or radiotherapy Download PDFInfo
- Publication number
- WO2024133723A1 WO2024133723A1 PCT/EP2023/087320 EP2023087320W WO2024133723A1 WO 2024133723 A1 WO2024133723 A1 WO 2024133723A1 EP 2023087320 W EP2023087320 W EP 2023087320W WO 2024133723 A1 WO2024133723 A1 WO 2024133723A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- tau
- cancer
- dna
- cells
- inhibitor
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims abstract description 51
- 238000001959 radiotherapy Methods 0.000 title claims abstract description 34
- 230000001225 therapeutic effect Effects 0.000 title claims abstract description 17
- 230000003247 decreasing effect Effects 0.000 title claims abstract description 10
- 238000002512 chemotherapy Methods 0.000 title abstract description 10
- 102000013498 tau Proteins Human genes 0.000 claims abstract description 205
- 108010026424 tau Proteins Proteins 0.000 claims abstract description 205
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 74
- 239000003112 inhibitor Substances 0.000 claims abstract description 56
- 238000011282 treatment Methods 0.000 claims abstract description 39
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin 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(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 claims abstract description 36
- 201000011510 cancer Diseases 0.000 claims abstract description 35
- 230000014509 gene expression Effects 0.000 claims abstract description 33
- 239000004055 small Interfering RNA Substances 0.000 claims abstract description 31
- 108091027967 Small hairpin RNA Proteins 0.000 claims abstract description 18
- 229960004679 doxorubicin Drugs 0.000 claims abstract description 18
- 239000003795 chemical substances by application Substances 0.000 claims abstract description 15
- 239000012829 chemotherapy agent Substances 0.000 claims abstract description 6
- 230000000694 effects Effects 0.000 claims description 30
- 239000002246 antineoplastic agent Substances 0.000 claims description 18
- 229960005486 vaccine Drugs 0.000 claims description 17
- 108020004459 Small interfering RNA Proteins 0.000 claims description 16
- 229960001756 oxaliplatin Drugs 0.000 claims description 15
- DWAFYCQODLXJNR-BNTLRKBRSA-L oxaliplatin Chemical compound O1C(=O)C(=O)O[Pt]11N[C@@H]2CCCC[C@H]2N1 DWAFYCQODLXJNR-BNTLRKBRSA-L 0.000 claims description 15
- 108091023037 Aptamer Proteins 0.000 claims description 13
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 claims description 13
- 229960004316 cisplatin Drugs 0.000 claims description 13
- 108090000994 Catalytic RNA Proteins 0.000 claims description 12
- 102000053642 Catalytic RNA Human genes 0.000 claims description 12
- 108091092562 ribozyme Proteins 0.000 claims description 12
- 108010026668 snake venom protein C activator Proteins 0.000 claims description 10
- 206010006187 Breast cancer Diseases 0.000 claims description 9
- 108091034117 Oligonucleotide Proteins 0.000 claims description 9
- 229940124823 proteolysis targeting chimeric molecule Drugs 0.000 claims description 9
- 208000026310 Breast neoplasm Diseases 0.000 claims description 8
- 229960004562 carboplatin Drugs 0.000 claims description 8
- 150000007523 nucleic acids Chemical group 0.000 claims description 8
- 238000011865 proteolysis targeting chimera technique Methods 0.000 claims description 8
- 206010029260 Neuroblastoma Diseases 0.000 claims description 6
- 206010060862 Prostate cancer Diseases 0.000 claims description 6
- 208000000236 Prostatic Neoplasms Diseases 0.000 claims description 6
- 208000032612 Glial tumor Diseases 0.000 claims description 5
- 206010018338 Glioma Diseases 0.000 claims description 5
- 101710163270 Nuclease Proteins 0.000 claims description 5
- 206010033128 Ovarian cancer Diseases 0.000 claims description 5
- 206010061535 Ovarian neoplasm Diseases 0.000 claims description 5
- 102000006275 Ubiquitin-Protein Ligases Human genes 0.000 claims description 5
- 108010083111 Ubiquitin-Protein Ligases Proteins 0.000 claims description 5
- 229940127089 cytotoxic agent Drugs 0.000 claims description 4
- 201000011519 neuroendocrine tumor Diseases 0.000 claims description 4
- 101000854943 Enterobacteria phage T4 Valyl-tRNA ligase modifier Proteins 0.000 claims description 3
- 108091028043 Nucleic acid sequence Proteins 0.000 claims description 3
- 230000000692 anti-sense effect Effects 0.000 claims description 3
- 208000030808 Clear cell renal carcinoma Diseases 0.000 claims description 2
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 2
- 206010061332 Paraganglion neoplasm Diseases 0.000 claims description 2
- 230000002496 gastric effect Effects 0.000 claims description 2
- 201000005202 lung cancer Diseases 0.000 claims description 2
- 208000020816 lung neoplasm Diseases 0.000 claims description 2
- 208000007312 paraganglioma Diseases 0.000 claims description 2
- 208000028591 pheochromocytoma Diseases 0.000 claims description 2
- 190000008236 carboplatin Chemical compound 0.000 claims 1
- 230000003439 radiotherapeutic effect Effects 0.000 claims 1
- 230000001965 increasing effect Effects 0.000 abstract description 17
- 230000001939 inductive effect Effects 0.000 abstract description 8
- 238000001727 in vivo Methods 0.000 abstract description 7
- 101150070547 MAPT gene Proteins 0.000 abstract description 3
- 230000035945 sensitivity Effects 0.000 abstract description 3
- 238000011275 oncology therapy Methods 0.000 abstract description 2
- 230000009467 reduction Effects 0.000 abstract description 2
- 210000004027 cell Anatomy 0.000 description 133
- 108090000623 proteins and genes Proteins 0.000 description 73
- 102000004169 proteins and genes Human genes 0.000 description 51
- 102000000504 Tumor Suppressor p53-Binding Protein 1 Human genes 0.000 description 50
- 108010041385 Tumor Suppressor p53-Binding Protein 1 Proteins 0.000 description 50
- 108020004414 DNA Proteins 0.000 description 40
- 230000033616 DNA repair Effects 0.000 description 36
- 230000008439 repair process Effects 0.000 description 36
- 238000002744 homologous recombination Methods 0.000 description 35
- 230000006801 homologous recombination Effects 0.000 description 35
- 102000029749 Microtubule Human genes 0.000 description 32
- 108091022875 Microtubule Proteins 0.000 description 32
- 210000004688 microtubule Anatomy 0.000 description 32
- 239000013612 plasmid Substances 0.000 description 29
- 230000006780 non-homologous end joining Effects 0.000 description 24
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 22
- 239000003814 drug Substances 0.000 description 19
- 230000027455 binding Effects 0.000 description 18
- 108010029485 Protein Isoforms Proteins 0.000 description 17
- 102000001708 Protein Isoforms Human genes 0.000 description 17
- 201000010099 disease Diseases 0.000 description 17
- 230000037361 pathway Effects 0.000 description 17
- 229940079593 drug Drugs 0.000 description 16
- 230000005855 radiation Effects 0.000 description 16
- WYWHKKSPHMUBEB-UHFFFAOYSA-N tioguanine Chemical compound N1C(N)=NC(=S)C2=C1N=CN2 WYWHKKSPHMUBEB-UHFFFAOYSA-N 0.000 description 16
- 238000002474 experimental method Methods 0.000 description 14
- 108010042407 Endonucleases Proteins 0.000 description 13
- 239000000203 mixture Substances 0.000 description 13
- 239000013598 vector Substances 0.000 description 13
- 102000004533 Endonucleases Human genes 0.000 description 12
- 238000004519 manufacturing process Methods 0.000 description 12
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 11
- 150000001875 compounds Chemical class 0.000 description 11
- 239000012634 fragment Substances 0.000 description 11
- 239000000427 antigen Substances 0.000 description 10
- 108091007433 antigens Proteins 0.000 description 10
- 102000036639 antigens Human genes 0.000 description 10
- 230000006698 induction Effects 0.000 description 10
- 230000003993 interaction Effects 0.000 description 10
- 108020004999 messenger RNA Proteins 0.000 description 10
- 210000004940 nucleus Anatomy 0.000 description 10
- 108090000765 processed proteins & peptides Proteins 0.000 description 10
- 238000012360 testing method Methods 0.000 description 10
- -1 ATR Proteins 0.000 description 9
- 230000005778 DNA damage Effects 0.000 description 9
- 231100000277 DNA damage Toxicity 0.000 description 9
- 229940123237 Taxane Drugs 0.000 description 9
- 230000001086 cytosolic effect Effects 0.000 description 9
- 230000005782 double-strand break Effects 0.000 description 9
- 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 9
- 230000005945 translocation Effects 0.000 description 9
- 230000032258 transport Effects 0.000 description 9
- 206010009944 Colon cancer Diseases 0.000 description 8
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 8
- 101150003028 Hprt1 gene Proteins 0.000 description 8
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 8
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 8
- 241001465754 Metazoa Species 0.000 description 8
- 241000699670 Mus sp. Species 0.000 description 8
- 230000007423 decrease Effects 0.000 description 8
- 102000013035 dynein heavy chain Human genes 0.000 description 8
- 108060002430 dynein heavy chain Proteins 0.000 description 8
- 229960002949 fluorouracil Drugs 0.000 description 8
- 230000000977 initiatory effect Effects 0.000 description 8
- 230000005865 ionizing radiation Effects 0.000 description 8
- 201000001441 melanoma Diseases 0.000 description 8
- 230000005937 nuclear translocation Effects 0.000 description 8
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 8
- 229960003087 tioguanine Drugs 0.000 description 8
- 210000001519 tissue Anatomy 0.000 description 8
- 108091033409 CRISPR Proteins 0.000 description 7
- 241000702421 Dependoparvovirus Species 0.000 description 7
- 241000700605 Viruses Species 0.000 description 7
- 239000000074 antisense oligonucleotide Substances 0.000 description 7
- 238000012230 antisense oligonucleotides Methods 0.000 description 7
- 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 7
- 230000004071 biological effect Effects 0.000 description 7
- 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 7
- 230000015556 catabolic process Effects 0.000 description 7
- 238000003776 cleavage reaction Methods 0.000 description 7
- 238000006731 degradation reaction Methods 0.000 description 7
- 230000006870 function Effects 0.000 description 7
- 230000005764 inhibitory process Effects 0.000 description 7
- 238000012423 maintenance Methods 0.000 description 7
- 239000008194 pharmaceutical composition Substances 0.000 description 7
- 230000007017 scission Effects 0.000 description 7
- 208000024891 symptom Diseases 0.000 description 7
- DKPFODGZWDEEBT-QFIAKTPHSA-N taxane Chemical class C([C@]1(C)CCC[C@@H](C)[C@H]1C1)C[C@H]2[C@H](C)CC[C@@H]1C2(C)C DKPFODGZWDEEBT-QFIAKTPHSA-N 0.000 description 7
- 241001430294 unidentified retrovirus Species 0.000 description 7
- 108010006654 Bleomycin Proteins 0.000 description 6
- 239000012623 DNA damaging agent Substances 0.000 description 6
- 101000891579 Homo sapiens Microtubule-associated protein tau Proteins 0.000 description 6
- 108060003951 Immunoglobulin Proteins 0.000 description 6
- 102100040243 Microtubule-associated protein tau Human genes 0.000 description 6
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 6
- 208000005718 Stomach Neoplasms Diseases 0.000 description 6
- 206010017758 gastric cancer Diseases 0.000 description 6
- 102000057063 human MAPT Human genes 0.000 description 6
- 102000018358 immunoglobulin Human genes 0.000 description 6
- 230000002401 inhibitory effect Effects 0.000 description 6
- 230000003902 lesion Effects 0.000 description 6
- 230000001537 neural effect Effects 0.000 description 6
- 239000002953 phosphate buffered saline Substances 0.000 description 6
- 238000010384 proximity ligation assay Methods 0.000 description 6
- 230000001105 regulatory effect Effects 0.000 description 6
- 230000004044 response Effects 0.000 description 6
- 150000003839 salts Chemical class 0.000 description 6
- 239000000243 solution Substances 0.000 description 6
- 201000011549 stomach cancer Diseases 0.000 description 6
- 239000000126 substance Substances 0.000 description 6
- 238000011285 therapeutic regimen Methods 0.000 description 6
- 238000001262 western blot Methods 0.000 description 6
- 208000024827 Alzheimer disease Diseases 0.000 description 5
- 208000001333 Colorectal Neoplasms Diseases 0.000 description 5
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 5
- 208000008839 Kidney Neoplasms Diseases 0.000 description 5
- 206010025323 Lymphomas Diseases 0.000 description 5
- 101710115937 Microtubule-associated protein tau Proteins 0.000 description 5
- 229930012538 Paclitaxel Natural products 0.000 description 5
- 206010038389 Renal cancer Diseases 0.000 description 5
- 206010039491 Sarcoma Diseases 0.000 description 5
- 239000002256 antimetabolite Substances 0.000 description 5
- 229960001561 bleomycin Drugs 0.000 description 5
- 210000000481 breast Anatomy 0.000 description 5
- 230000006378 damage Effects 0.000 description 5
- 208000035475 disorder Diseases 0.000 description 5
- 238000005516 engineering process Methods 0.000 description 5
- 201000010982 kidney cancer Diseases 0.000 description 5
- 230000007246 mechanism Effects 0.000 description 5
- GLVAUDGFNGKCSF-UHFFFAOYSA-N mercaptopurine Chemical compound S=C1NC=NC2=C1NC=N2 GLVAUDGFNGKCSF-UHFFFAOYSA-N 0.000 description 5
- 230000008880 microtubule cytoskeleton organization Effects 0.000 description 5
- 230000035772 mutation Effects 0.000 description 5
- 229960001592 paclitaxel Drugs 0.000 description 5
- 238000002360 preparation method Methods 0.000 description 5
- 102000004196 processed proteins & peptides Human genes 0.000 description 5
- 238000002560 therapeutic procedure Methods 0.000 description 5
- 239000003981 vehicle Substances 0.000 description 5
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 description 4
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 description 4
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 4
- 241000894006 Bacteria Species 0.000 description 4
- 238000010354 CRISPR gene editing Methods 0.000 description 4
- GAGWJHPBXLXJQN-UORFTKCHSA-N Capecitabine Chemical compound C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1[C@H]1[C@H](O)[C@H](O)[C@@H](C)O1 GAGWJHPBXLXJQN-UORFTKCHSA-N 0.000 description 4
- 206010008342 Cervix carcinoma Diseases 0.000 description 4
- 239000004971 Cross linker Substances 0.000 description 4
- 206010014759 Endometrial neoplasm Diseases 0.000 description 4
- 102000004190 Enzymes Human genes 0.000 description 4
- 108090000790 Enzymes Proteins 0.000 description 4
- 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 4
- 241000124008 Mammalia Species 0.000 description 4
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 description 4
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 4
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 4
- 101710183280 Topoisomerase Proteins 0.000 description 4
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 description 4
- 150000001413 amino acids Chemical class 0.000 description 4
- 238000011394 anticancer treatment Methods 0.000 description 4
- 238000003556 assay Methods 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 230000001413 cellular effect Effects 0.000 description 4
- 201000010881 cervical cancer Diseases 0.000 description 4
- 238000012217 deletion Methods 0.000 description 4
- 230000037430 deletion Effects 0.000 description 4
- 239000006185 dispersion Substances 0.000 description 4
- 229940088598 enzyme Drugs 0.000 description 4
- 238000000684 flow cytometry Methods 0.000 description 4
- 238000003197 gene knockdown Methods 0.000 description 4
- 230000009368 gene silencing by RNA Effects 0.000 description 4
- 208000005017 glioblastoma Diseases 0.000 description 4
- 201000010536 head and neck cancer Diseases 0.000 description 4
- 208000014829 head and neck neoplasm Diseases 0.000 description 4
- 210000004408 hybridoma Anatomy 0.000 description 4
- FDGQSTZJBFJUBT-UHFFFAOYSA-N hypoxanthine Chemical compound O=C1NC=NC2=C1NC=N2 FDGQSTZJBFJUBT-UHFFFAOYSA-N 0.000 description 4
- 238000000338 in vitro Methods 0.000 description 4
- 208000032839 leukemia Diseases 0.000 description 4
- 230000000670 limiting effect Effects 0.000 description 4
- 210000004962 mammalian cell Anatomy 0.000 description 4
- 229960000485 methotrexate Drugs 0.000 description 4
- KKZJGLLVHKMTCM-UHFFFAOYSA-N mitoxantrone Chemical compound O=C1C2=C(O)C=CC(O)=C2C(=O)C2=C1C(NCCNCCO)=CC=C2NCCNCCO KKZJGLLVHKMTCM-UHFFFAOYSA-N 0.000 description 4
- 230000036438 mutation frequency Effects 0.000 description 4
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 4
- 102000039446 nucleic acids Human genes 0.000 description 4
- 108020004707 nucleic acids Proteins 0.000 description 4
- 230000006849 nucleocytoplasmic transport Effects 0.000 description 4
- BWKDAMBGCPRVPI-ZQRPHVBESA-N ortataxel Chemical compound O([C@@H]1[C@]23OC(=O)O[C@H]2[C@@H](C(=C([C@@H](OC(C)=O)C(=O)[C@]2(C)[C@@H](O)C[C@H]4OC[C@]4([C@H]21)OC(C)=O)C3(C)C)C)OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)CC(C)C)C(=O)C1=CC=CC=C1 BWKDAMBGCPRVPI-ZQRPHVBESA-N 0.000 description 4
- 239000002245 particle Substances 0.000 description 4
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 4
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 4
- 229920001184 polypeptide Polymers 0.000 description 4
- 229950010131 puromycin Drugs 0.000 description 4
- 230000002829 reductive effect Effects 0.000 description 4
- 239000011780 sodium chloride Substances 0.000 description 4
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 4
- 230000008685 targeting Effects 0.000 description 4
- 210000001685 thyroid gland Anatomy 0.000 description 4
- 231100000331 toxic Toxicity 0.000 description 4
- 230000002588 toxic effect Effects 0.000 description 4
- 230000014616 translation Effects 0.000 description 4
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 description 4
- FDKXTQMXEQVLRF-ZHACJKMWSA-N (E)-dacarbazine Chemical compound CN(C)\N=N\c1[nH]cnc1C(N)=O FDKXTQMXEQVLRF-ZHACJKMWSA-N 0.000 description 3
- AOJJSUZBOXZQNB-VTZDEGQISA-N 4'-epidoxorubicin 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(=O)CO)[C@H]1C[C@H](N)[C@@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-VTZDEGQISA-N 0.000 description 3
- 208000031261 Acute myeloid leukaemia Diseases 0.000 description 3
- 108010012934 Albumin-Bound Paclitaxel Proteins 0.000 description 3
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 3
- 108700020463 BRCA1 Proteins 0.000 description 3
- 102000036365 BRCA1 Human genes 0.000 description 3
- 206010005003 Bladder cancer Diseases 0.000 description 3
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 3
- KLWPJMFMVPTNCC-UHFFFAOYSA-N Camptothecin Natural products CCC1(O)C(=O)OCC2=C1C=C3C4Nc5ccccc5C=C4CN3C2=O KLWPJMFMVPTNCC-UHFFFAOYSA-N 0.000 description 3
- GAGWJHPBXLXJQN-UHFFFAOYSA-N Capecitabine Natural products C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1C1C(O)C(O)C(C)O1 GAGWJHPBXLXJQN-UHFFFAOYSA-N 0.000 description 3
- DLGOEMSEDOSKAD-UHFFFAOYSA-N Carmustine Chemical compound ClCCNC(=O)N(N=O)CCCl DLGOEMSEDOSKAD-UHFFFAOYSA-N 0.000 description 3
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 3
- 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 3
- 241000196324 Embryophyta Species 0.000 description 3
- 241000588724 Escherichia coli Species 0.000 description 3
- 206010017993 Gastrointestinal neoplasms Diseases 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 102100034533 Histone H2AX Human genes 0.000 description 3
- 101001067891 Homo sapiens Histone H2AX Proteins 0.000 description 3
- 201000003793 Myelodysplastic syndrome Diseases 0.000 description 3
- 208000033776 Myeloid Acute Leukemia Diseases 0.000 description 3
- 241000283973 Oryctolagus cuniculus Species 0.000 description 3
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 3
- 241000288906 Primates Species 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 208000019155 Radiation injury Diseases 0.000 description 3
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 3
- 206010041067 Small cell lung cancer Diseases 0.000 description 3
- FOCVUCIESVLUNU-UHFFFAOYSA-N Thiotepa Chemical compound C1CN1P(N1CC1)(=S)N1CC1 FOCVUCIESVLUNU-UHFFFAOYSA-N 0.000 description 3
- 208000024770 Thyroid neoplasm Diseases 0.000 description 3
- 239000013504 Triton X-100 Substances 0.000 description 3
- 229920004890 Triton X-100 Polymers 0.000 description 3
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 description 3
- 229940028652 abraxane Drugs 0.000 description 3
- 239000002253 acid Substances 0.000 description 3
- 230000009471 action Effects 0.000 description 3
- 239000004480 active ingredient Substances 0.000 description 3
- 101150063416 add gene Proteins 0.000 description 3
- 230000002411 adverse Effects 0.000 description 3
- 230000004075 alteration Effects 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- 210000003719 b-lymphocyte Anatomy 0.000 description 3
- 230000008901 benefit Effects 0.000 description 3
- 210000004556 brain Anatomy 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 229930195731 calicheamicin Natural products 0.000 description 3
- HXCHCVDVKSCDHU-LULTVBGHSA-N calicheamicin Chemical compound C1[C@H](OC)[C@@H](NCC)CO[C@H]1O[C@H]1[C@H](O[C@@H]2C\3=C(NC(=O)OC)C(=O)C[C@](C/3=C/CSSSC)(O)C#C\C=C/C#C2)O[C@H](C)[C@@H](NO[C@@H]2O[C@H](C)[C@@H](SC(=O)C=3C(=C(OC)C(O[C@H]4[C@@H]([C@H](OC)[C@@H](O)[C@H](C)O4)O)=C(I)C=3C)OC)[C@@H](O)C2)[C@@H]1O HXCHCVDVKSCDHU-LULTVBGHSA-N 0.000 description 3
- VSJKWCGYPAHWDS-FQEVSTJZSA-N camptothecin Chemical compound C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-FQEVSTJZSA-N 0.000 description 3
- 229940127093 camptothecin Drugs 0.000 description 3
- 229960004117 capecitabine Drugs 0.000 description 3
- 229960005243 carmustine Drugs 0.000 description 3
- 210000003679 cervix uteri Anatomy 0.000 description 3
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 3
- 229960004630 chlorambucil Drugs 0.000 description 3
- JCKYGMPEJWAADB-UHFFFAOYSA-N chlorambucil Chemical compound OC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 JCKYGMPEJWAADB-UHFFFAOYSA-N 0.000 description 3
- 230000000295 complement effect Effects 0.000 description 3
- 229960003901 dacarbazine Drugs 0.000 description 3
- 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 3
- 230000002950 deficient Effects 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 239000000539 dimer Substances 0.000 description 3
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 3
- 238000009826 distribution Methods 0.000 description 3
- VHJLVAABSRFDPM-QWWZWVQMSA-N dithiothreitol Chemical compound SC[C@@H](O)[C@H](O)CS VHJLVAABSRFDPM-QWWZWVQMSA-N 0.000 description 3
- VSJKWCGYPAHWDS-UHFFFAOYSA-N dl-camptothecin Natural products C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)C5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-UHFFFAOYSA-N 0.000 description 3
- 239000003937 drug carrier Substances 0.000 description 3
- 210000003238 esophagus Anatomy 0.000 description 3
- 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 3
- 239000000284 extract Substances 0.000 description 3
- 239000012091 fetal bovine serum Substances 0.000 description 3
- 238000009472 formulation Methods 0.000 description 3
- 238000005194 fractionation Methods 0.000 description 3
- 229960005277 gemcitabine Drugs 0.000 description 3
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 3
- 201000005787 hematologic cancer Diseases 0.000 description 3
- 208000024200 hematopoietic and lymphoid system neoplasm Diseases 0.000 description 3
- 229960001101 ifosfamide Drugs 0.000 description 3
- HOMGKSMUEGBAAB-UHFFFAOYSA-N ifosfamide Chemical compound ClCCNP1(=O)OCCCN1CCCl HOMGKSMUEGBAAB-UHFFFAOYSA-N 0.000 description 3
- 238000002649 immunization Methods 0.000 description 3
- 230000003053 immunization Effects 0.000 description 3
- 239000004615 ingredient Substances 0.000 description 3
- 238000007918 intramuscular administration Methods 0.000 description 3
- 210000003734 kidney Anatomy 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 210000004185 liver Anatomy 0.000 description 3
- 210000004072 lung Anatomy 0.000 description 3
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 229960001428 mercaptopurine Drugs 0.000 description 3
- 229960001156 mitoxantrone Drugs 0.000 description 3
- 230000004770 neurodegeneration Effects 0.000 description 3
- 210000002569 neuron Anatomy 0.000 description 3
- 230000020520 nucleotide-excision repair Effects 0.000 description 3
- 210000001672 ovary Anatomy 0.000 description 3
- 230000002018 overexpression Effects 0.000 description 3
- 210000000496 pancreas Anatomy 0.000 description 3
- 201000002528 pancreatic cancer Diseases 0.000 description 3
- 208000008443 pancreatic carcinoma Diseases 0.000 description 3
- 230000001575 pathological effect Effects 0.000 description 3
- 239000000546 pharmaceutical excipient Substances 0.000 description 3
- 239000013600 plasmid vector Substances 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 210000002307 prostate Anatomy 0.000 description 3
- 230000004224 protection Effects 0.000 description 3
- 230000001681 protective effect Effects 0.000 description 3
- 238000011002 quantification Methods 0.000 description 3
- 230000007115 recruitment Effects 0.000 description 3
- 230000010076 replication Effects 0.000 description 3
- 239000006152 selective media Substances 0.000 description 3
- 208000000587 small cell lung carcinoma Diseases 0.000 description 3
- 239000007787 solid Substances 0.000 description 3
- 239000002904 solvent Substances 0.000 description 3
- 206010041823 squamous cell carcinoma Diseases 0.000 description 3
- 238000007920 subcutaneous administration Methods 0.000 description 3
- 229960001196 thiotepa Drugs 0.000 description 3
- 201000002510 thyroid cancer Diseases 0.000 description 3
- 229960000303 topotecan Drugs 0.000 description 3
- UCFGDBYHRUNTLO-QHCPKHFHSA-N topotecan Chemical compound C1=C(O)C(CN(C)C)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 UCFGDBYHRUNTLO-QHCPKHFHSA-N 0.000 description 3
- 238000010361 transduction Methods 0.000 description 3
- 230000026683 transduction Effects 0.000 description 3
- 238000001890 transfection Methods 0.000 description 3
- 238000013519 translation Methods 0.000 description 3
- 210000003932 urinary bladder Anatomy 0.000 description 3
- 201000005112 urinary bladder cancer Diseases 0.000 description 3
- 239000013603 viral vector Substances 0.000 description 3
- 230000003612 virological effect Effects 0.000 description 3
- MQLACMBJVPINKE-UHFFFAOYSA-N 10-[(3-hydroxy-4-methoxyphenyl)methylidene]anthracen-9-one Chemical compound C1=C(O)C(OC)=CC=C1C=C1C2=CC=CC=C2C(=O)C2=CC=CC=C21 MQLACMBJVPINKE-UHFFFAOYSA-N 0.000 description 2
- 102100027831 14-3-3 protein theta Human genes 0.000 description 2
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 2
- IDPUKCWIGUEADI-UHFFFAOYSA-N 5-[bis(2-chloroethyl)amino]uracil Chemical compound ClCCN(CCCl)C1=CNC(=O)NC1=O IDPUKCWIGUEADI-UHFFFAOYSA-N 0.000 description 2
- 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 2
- 108700028369 Alleles Proteins 0.000 description 2
- 108020005544 Antisense RNA Proteins 0.000 description 2
- 108010004586 Ataxia Telangiectasia Mutated Proteins Proteins 0.000 description 2
- NOWKCMXCCJGMRR-UHFFFAOYSA-N Aziridine Chemical class C1CN1 NOWKCMXCCJGMRR-UHFFFAOYSA-N 0.000 description 2
- 208000032791 BCR-ABL1 positive chronic myelogenous leukemia Diseases 0.000 description 2
- 208000018084 Bone neoplasm Diseases 0.000 description 2
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 2
- 208000005623 Carcinogenesis Diseases 0.000 description 2
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 2
- 102000053602 DNA Human genes 0.000 description 2
- 239000012624 DNA alkylating agent Substances 0.000 description 2
- 102100022204 DNA-dependent protein kinase catalytic subunit Human genes 0.000 description 2
- 101710157074 DNA-dependent protein kinase catalytic subunit Proteins 0.000 description 2
- 108010092160 Dactinomycin Proteins 0.000 description 2
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 2
- 206010014733 Endometrial cancer Diseases 0.000 description 2
- HTIJFSOGRVMCQR-UHFFFAOYSA-N Epirubicin Natural products COc1cccc2C(=O)c3c(O)c4CC(O)(CC(OC5CC(N)C(=O)C(C)O5)c4c(O)c3C(=O)c12)C(=O)CO HTIJFSOGRVMCQR-UHFFFAOYSA-N 0.000 description 2
- 201000008808 Fibrosarcoma Diseases 0.000 description 2
- 201000011240 Frontotemporal dementia Diseases 0.000 description 2
- 208000021309 Germ cell tumor Diseases 0.000 description 2
- 201000010915 Glioblastoma multiforme Diseases 0.000 description 2
- 101000721661 Homo sapiens Cellular tumor antigen p53 Proteins 0.000 description 2
- 102000018251 Hypoxanthine Phosphoribosyltransferase Human genes 0.000 description 2
- 108010091358 Hypoxanthine Phosphoribosyltransferase Proteins 0.000 description 2
- UGQMRVRMYYASKQ-UHFFFAOYSA-N Hypoxanthine nucleoside Natural products OC1C(O)C(CO)OC1N1C(NC=NC2=O)=C2N=C1 UGQMRVRMYYASKQ-UHFFFAOYSA-N 0.000 description 2
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 2
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 2
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 2
- 229930182816 L-glutamine Natural products 0.000 description 2
- GQYIWUVLTXOXAJ-UHFFFAOYSA-N Lomustine Chemical compound ClCCN(N=O)C(=O)NC1CCCCC1 GQYIWUVLTXOXAJ-UHFFFAOYSA-N 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- 238000000585 Mann–Whitney U test Methods 0.000 description 2
- 102000009664 Microtubule-Associated Proteins Human genes 0.000 description 2
- 108010020004 Microtubule-Associated Proteins Proteins 0.000 description 2
- 208000033761 Myelogenous Chronic BCR-ABL Positive Leukemia Diseases 0.000 description 2
- 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 2
- 108091007491 NSP3 Papain-like protease domains Proteins 0.000 description 2
- 208000034176 Neoplasms, Germ Cell and Embryonal Diseases 0.000 description 2
- 229930182555 Penicillin Natural products 0.000 description 2
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 2
- 102000057297 Pepsin A Human genes 0.000 description 2
- 108090000284 Pepsin A Proteins 0.000 description 2
- 241000235648 Pichia Species 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- 102000000574 RNA-Induced Silencing Complex Human genes 0.000 description 2
- 108010016790 RNA-Induced Silencing Complex Proteins 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 108700008625 Reporter Genes Proteins 0.000 description 2
- 241000283984 Rodentia Species 0.000 description 2
- 238000011579 SCID mouse model Methods 0.000 description 2
- 241000282887 Suidae Species 0.000 description 2
- 210000001744 T-lymphocyte Anatomy 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
- 102000004243 Tubulin Human genes 0.000 description 2
- 108090000704 Tubulin Proteins 0.000 description 2
- 108090000848 Ubiquitin Proteins 0.000 description 2
- 102000044159 Ubiquitin Human genes 0.000 description 2
- 150000007513 acids Chemical class 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
- 210000000577 adipose tissue Anatomy 0.000 description 2
- 229940045714 alkyl sulfonate alkylating agent Drugs 0.000 description 2
- 150000008052 alkyl sulfonates Chemical class 0.000 description 2
- 229940100198 alkylating agent Drugs 0.000 description 2
- 239000002168 alkylating agent Substances 0.000 description 2
- 229960001220 amsacrine Drugs 0.000 description 2
- XCPGHVQEEXUHNC-UHFFFAOYSA-N amsacrine Chemical compound COC1=CC(NS(C)(=O)=O)=CC=C1NC1=C(C=CC=C2)C2=NC2=CC=CC=C12 XCPGHVQEEXUHNC-UHFFFAOYSA-N 0.000 description 2
- 239000005557 antagonist Substances 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 229940088710 antibiotic agent Drugs 0.000 description 2
- 229940041181 antineoplastic drug Drugs 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 239000007864 aqueous solution Substances 0.000 description 2
- 210000003050 axon Anatomy 0.000 description 2
- 230000001580 bacterial effect Effects 0.000 description 2
- 230000033590 base-excision repair Effects 0.000 description 2
- 230000009286 beneficial effect Effects 0.000 description 2
- 230000001588 bifunctional effect Effects 0.000 description 2
- 229960002092 busulfan Drugs 0.000 description 2
- 229960001573 cabazitaxel Drugs 0.000 description 2
- BMQGVNUXMIRLCK-OAGWZNDDSA-N cabazitaxel Chemical compound O([C@H]1[C@@H]2[C@]3(OC(C)=O)CO[C@@H]3C[C@@H]([C@]2(C(=O)[C@H](OC)C2=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=3C=CC=CC=3)C[C@]1(O)C2(C)C)C)OC)C(=O)C1=CC=CC=C1 BMQGVNUXMIRLCK-OAGWZNDDSA-N 0.000 description 2
- 230000036952 cancer formation Effects 0.000 description 2
- 231100000504 carcinogenesis Toxicity 0.000 description 2
- 239000000969 carrier Substances 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 230000022131 cell cycle Effects 0.000 description 2
- 230000030833 cell death Effects 0.000 description 2
- 108091092356 cellular DNA Proteins 0.000 description 2
- 230000005754 cellular signaling Effects 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- 210000000349 chromosome Anatomy 0.000 description 2
- 238000004891 communication Methods 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 239000003184 complementary RNA Substances 0.000 description 2
- 229960004397 cyclophosphamide Drugs 0.000 description 2
- 229960000684 cytarabine Drugs 0.000 description 2
- 210000000805 cytoplasm Anatomy 0.000 description 2
- 231100000433 cytotoxic Toxicity 0.000 description 2
- 230000001472 cytotoxic effect Effects 0.000 description 2
- 230000000254 damaging effect Effects 0.000 description 2
- 229960000975 daunorubicin Drugs 0.000 description 2
- 230000007547 defect Effects 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000029087 digestion Effects 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 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 2
- 230000034431 double-strand break repair via homologous recombination Effects 0.000 description 2
- 230000009977 dual effect Effects 0.000 description 2
- VLCYCQAOQCDTCN-UHFFFAOYSA-N eflornithine Chemical compound NCCCC(N)(C(F)F)C(O)=O VLCYCQAOQCDTCN-UHFFFAOYSA-N 0.000 description 2
- 230000002357 endometrial effect Effects 0.000 description 2
- 230000002616 endonucleolytic effect Effects 0.000 description 2
- 229960001904 epirubicin Drugs 0.000 description 2
- DEFVIWRASFVYLL-UHFFFAOYSA-N ethylene glycol bis(2-aminoethyl)tetraacetic acid Chemical compound OC(=O)CN(CC(O)=O)CCOCCOCCN(CC(O)=O)CC(O)=O DEFVIWRASFVYLL-UHFFFAOYSA-N 0.000 description 2
- 229960005420 etoposide Drugs 0.000 description 2
- 238000011347 external beam therapy Methods 0.000 description 2
- 229960000961 floxuridine Drugs 0.000 description 2
- ODKNJVUHOIMIIZ-RRKCRQDMSA-N floxuridine Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(F)=C1 ODKNJVUHOIMIIZ-RRKCRQDMSA-N 0.000 description 2
- GIUYCYHIANZCFB-FJFJXFQQSA-N fludarabine phosphate Chemical compound C1=NC=2C(N)=NC(F)=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@@H]1O GIUYCYHIANZCFB-FJFJXFQQSA-N 0.000 description 2
- OVBPIULPVIDEAO-LBPRGKRZSA-N folic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-LBPRGKRZSA-N 0.000 description 2
- 229960004783 fotemustine Drugs 0.000 description 2
- YAKWPXVTIGTRJH-UHFFFAOYSA-N fotemustine Chemical compound CCOP(=O)(OCC)C(C)NC(=O)N(CCCl)N=O YAKWPXVTIGTRJH-UHFFFAOYSA-N 0.000 description 2
- 230000004927 fusion Effects 0.000 description 2
- CHPZKNULDCNCBW-UHFFFAOYSA-N gallium nitrate Chemical compound [Ga+3].[O-][N+]([O-])=O.[O-][N+]([O-])=O.[O-][N+]([O-])=O CHPZKNULDCNCBW-UHFFFAOYSA-N 0.000 description 2
- 230000030279 gene silencing Effects 0.000 description 2
- 210000003128 head Anatomy 0.000 description 2
- 238000009396 hybridization Methods 0.000 description 2
- 238000002786 image-guided radiation therapy Methods 0.000 description 2
- 230000028993 immune response Effects 0.000 description 2
- 230000016784 immunoglobulin production Effects 0.000 description 2
- 230000000415 inactivating effect Effects 0.000 description 2
- 238000010348 incorporation Methods 0.000 description 2
- 208000015181 infectious disease Diseases 0.000 description 2
- 238000003780 insertion Methods 0.000 description 2
- 230000037431 insertion Effects 0.000 description 2
- 230000010354 integration Effects 0.000 description 2
- 238000002721 intensity-modulated radiation therapy Methods 0.000 description 2
- 230000003834 intracellular effect Effects 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- UWKQSNNFCGGAFS-XIFFEERXSA-N irinotecan Chemical compound C1=C2C(CC)=C3CN(C(C4=C([C@@](C(=O)OC4)(O)CC)C=4)=O)C=4C3=NC2=CC=C1OC(=O)N(CC1)CCC1N1CCCCC1 UWKQSNNFCGGAFS-XIFFEERXSA-N 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- 108010045069 keyhole-limpet hemocyanin Proteins 0.000 description 2
- DXOJIXGRFSHVKA-BZVZGCBYSA-N larotaxel Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@@]23[C@H]1[C@@]1(CO[C@@H]1C[C@@H]2C3)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 DXOJIXGRFSHVKA-BZVZGCBYSA-N 0.000 description 2
- 239000002502 liposome Substances 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 201000007270 liver cancer Diseases 0.000 description 2
- 208000014018 liver neoplasm Diseases 0.000 description 2
- 229960002247 lomustine Drugs 0.000 description 2
- 239000012139 lysis buffer Substances 0.000 description 2
- 208000008585 mastocytosis Diseases 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
- 229960001924 melphalan Drugs 0.000 description 2
- 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 2
- 230000003278 mimic effect Effects 0.000 description 2
- 230000033607 mismatch repair Effects 0.000 description 2
- 230000000394 mitotic effect Effects 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 230000009456 molecular mechanism Effects 0.000 description 2
- 231100000350 mutagenesis Toxicity 0.000 description 2
- 210000003739 neck Anatomy 0.000 description 2
- QZGIWPZCWHMVQL-UIYAJPBUSA-N neocarzinostatin chromophore Chemical compound O1[C@H](C)[C@H](O)[C@H](O)[C@@H](NC)[C@H]1O[C@@H]1C/2=C/C#C[C@H]3O[C@@]3([C@@H]3OC(=O)OC3)C#CC\2=C[C@H]1OC(=O)C1=C(O)C=CC2=C(C)C=C(OC)C=C12 QZGIWPZCWHMVQL-UIYAJPBUSA-N 0.000 description 2
- 229910052757 nitrogen Inorganic materials 0.000 description 2
- 210000000633 nuclear envelope Anatomy 0.000 description 2
- 230000030648 nucleus localization Effects 0.000 description 2
- 238000004806 packaging method and process Methods 0.000 description 2
- 238000004091 panning Methods 0.000 description 2
- 229940049954 penicillin Drugs 0.000 description 2
- FPVKHBSQESCIEP-JQCXWYLXSA-N pentostatin Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(N=CNC[C@H]2O)=C2N=C1 FPVKHBSQESCIEP-JQCXWYLXSA-N 0.000 description 2
- 229940111202 pepsin Drugs 0.000 description 2
- 201000002628 peritoneum cancer Diseases 0.000 description 2
- 229960000952 pipobroman Drugs 0.000 description 2
- NJBFOOCLYDNZJN-UHFFFAOYSA-N pipobroman Chemical compound BrCCC(=O)N1CCN(C(=O)CCBr)CC1 NJBFOOCLYDNZJN-UHFFFAOYSA-N 0.000 description 2
- 229910052697 platinum Inorganic materials 0.000 description 2
- 229920001223 polyethylene glycol Polymers 0.000 description 2
- 230000000069 prophylactic effect Effects 0.000 description 2
- 229940121649 protein inhibitor Drugs 0.000 description 2
- 239000012268 protein inhibitor Substances 0.000 description 2
- 230000026447 protein localization Effects 0.000 description 2
- 230000007398 protein translocation Effects 0.000 description 2
- 150000003212 purines Chemical class 0.000 description 2
- 150000003230 pyrimidines Chemical class 0.000 description 2
- 229960004622 raloxifene Drugs 0.000 description 2
- GZUITABIAKMVPG-UHFFFAOYSA-N raloxifene Chemical compound C1=CC(O)=CC=C1C1=C(C(=O)C=2C=CC(OCCN3CCCCC3)=CC=2)C2=CC=C(O)C=C2S1 GZUITABIAKMVPG-UHFFFAOYSA-N 0.000 description 2
- 230000008707 rearrangement Effects 0.000 description 2
- 230000006798 recombination Effects 0.000 description 2
- 238000005215 recombination Methods 0.000 description 2
- 230000028617 response to DNA damage stimulus Effects 0.000 description 2
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 2
- 239000001488 sodium phosphate Substances 0.000 description 2
- 230000002269 spontaneous effect Effects 0.000 description 2
- 229960005322 streptomycin Drugs 0.000 description 2
- 229960001052 streptozocin Drugs 0.000 description 2
- 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 2
- 239000000758 substrate Substances 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- MODVSQKJJIBWPZ-VLLPJHQWSA-N tesetaxel Chemical compound O([C@H]1[C@@H]2[C@]3(OC(C)=O)CO[C@@H]3CC[C@@]2(C)[C@H]2[C@@H](C3=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=4C(=CC=CN=4)F)C[C@]1(O)C3(C)C)O[C@H](O2)CN(C)C)C(=O)C1=CC=CC=C1 MODVSQKJJIBWPZ-VLLPJHQWSA-N 0.000 description 2
- 229950001353 tretamine Drugs 0.000 description 2
- IUCJMVBFZDHPDX-UHFFFAOYSA-N tretamine Chemical compound C1CN1C1=NC(N2CC2)=NC(N2CC2)=N1 IUCJMVBFZDHPDX-UHFFFAOYSA-N 0.000 description 2
- 230000004614 tumor growth Effects 0.000 description 2
- 241000701161 unidentified adenovirus Species 0.000 description 2
- 229960001055 uracil mustard Drugs 0.000 description 2
- 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 2
- 229960004528 vincristine Drugs 0.000 description 2
- 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 2
- GBABOYUKABKIAF-IELIFDKJSA-N vinorelbine Chemical compound C1N(CC=2C3=CC=CC=C3NC=22)CC(CC)=C[C@H]1C[C@]2(C(=O)OC)C1=CC([C@]23[C@H]([C@@]([C@H](OC(C)=O)[C@]4(CC)C=CCN([C@H]34)CC2)(O)C(=O)OC)N2C)=C2C=C1OC GBABOYUKABKIAF-IELIFDKJSA-N 0.000 description 2
- 229960002066 vinorelbine Drugs 0.000 description 2
- NNJPGOLRFBJNIW-HNNXBMFYSA-N (-)-demecolcine Chemical compound C1=C(OC)C(=O)C=C2[C@@H](NC)CCC3=CC(OC)=C(OC)C(OC)=C3C2=C1 NNJPGOLRFBJNIW-HNNXBMFYSA-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
- WDQLRUYAYXDIFW-RWKIJVEZSA-N (2r,3r,4s,5r,6r)-4-[(2s,3r,4s,5r,6r)-3,5-dihydroxy-4-[(2r,3r,4s,5s,6r)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-6-[[(2r,3r,4s,5s,6r)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxymethyl]oxan-2-yl]oxy-6-(hydroxymethyl)oxane-2,3,5-triol Chemical compound O[C@@H]1[C@@H](CO)O[C@@H](O)[C@H](O)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)O)[C@H](O)[C@@H](CO[C@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)O)O1 WDQLRUYAYXDIFW-RWKIJVEZSA-N 0.000 description 1
- FLWWDYNPWOSLEO-HQVZTVAUSA-N (2s)-2-[[4-[1-(2-amino-4-oxo-1h-pteridin-6-yl)ethyl-methylamino]benzoyl]amino]pentanedioic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1C(C)N(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FLWWDYNPWOSLEO-HQVZTVAUSA-N 0.000 description 1
- HONKEGXLWUDTCF-YFKPBYRVSA-N (2s)-2-amino-2-methyl-4-phosphonobutanoic acid Chemical compound OC(=O)[C@](N)(C)CCP(O)(O)=O HONKEGXLWUDTCF-YFKPBYRVSA-N 0.000 description 1
- CGMTUJFWROPELF-YPAAEMCBSA-N (3E,5S)-5-[(2S)-butan-2-yl]-3-(1-hydroxyethylidene)pyrrolidine-2,4-dione Chemical compound CC[C@H](C)[C@@H]1NC(=O)\C(=C(/C)O)C1=O CGMTUJFWROPELF-YPAAEMCBSA-N 0.000 description 1
- TVIRNGFXQVMMGB-OFWIHYRESA-N (3s,6r,10r,13e,16s)-16-[(2r,3r,4s)-4-chloro-3-hydroxy-4-phenylbutan-2-yl]-10-[(3-chloro-4-methoxyphenyl)methyl]-6-methyl-3-(2-methylpropyl)-1,4-dioxa-8,11-diazacyclohexadec-13-ene-2,5,9,12-tetrone 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](O)[C@@H](Cl)C=2C=CC=CC=2)C/C=C/C(=O)N1 TVIRNGFXQVMMGB-OFWIHYRESA-N 0.000 description 1
- XRBSKUSTLXISAB-XVVDYKMHSA-N (5r,6r,7r,8r)-8-hydroxy-7-(hydroxymethyl)-5-(3,4,5-trimethoxyphenyl)-5,6,7,8-tetrahydrobenzo[f][1,3]benzodioxole-6-carboxylic acid Chemical compound COC1=C(OC)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@H](O)[C@@H](CO)[C@@H]2C(O)=O)=C1 XRBSKUSTLXISAB-XVVDYKMHSA-N 0.000 description 1
- XRBSKUSTLXISAB-UHFFFAOYSA-N (7R,7'R,8R,8'R)-form-Podophyllic acid Natural products COC1=C(OC)C(OC)=CC(C2C3=CC=4OCOC=4C=C3C(O)C(CO)C2C(O)=O)=C1 XRBSKUSTLXISAB-UHFFFAOYSA-N 0.000 description 1
- AESVUZLWRXEGEX-DKCAWCKPSA-N (7S,9R)-7-[(2S,4R,5R,6R)-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 iron(3+) Chemical compound [Fe+3].COc1cccc2C(=O)c3c(O)c4C[C@@](O)(C[C@H](O[C@@H]5C[C@@H](N)[C@@H](O)[C@@H](C)O5)c4c(O)c3C(=O)c12)C(=O)CO AESVUZLWRXEGEX-DKCAWCKPSA-N 0.000 description 1
- JXVAMODRWBNUSF-KZQKBALLSA-N (7s,9r,10r)-7-[(2r,4s,5s,6s)-5-[[(2s,4as,5as,7s,9s,9ar,10ar)-2,9-dimethyl-3-oxo-4,4a,5a,6,7,9,9a,10a-octahydrodipyrano[4,2-a:4',3'-e][1,4]dioxin-7-yl]oxy]-4-(dimethylamino)-6-methyloxan-2-yl]oxy-10-[(2s,4s,5s,6s)-4-(dimethylamino)-5-hydroxy-6-methyloxan-2 Chemical compound O([C@@H]1C2=C(O)C=3C(=O)C4=CC=CC(O)=C4C(=O)C=3C(O)=C2[C@@H](O[C@@H]2O[C@@H](C)[C@@H](O[C@@H]3O[C@@H](C)[C@H]4O[C@@H]5O[C@@H](C)C(=O)C[C@@H]5O[C@H]4C3)[C@H](C2)N(C)C)C[C@]1(O)CC)[C@H]1C[C@H](N(C)C)[C@H](O)[C@H](C)O1 JXVAMODRWBNUSF-KZQKBALLSA-N 0.000 description 1
- INAUWOVKEZHHDM-PEDBPRJASA-N (7s,9s)-6,9,11-trihydroxy-9-(2-hydroxyacetyl)-7-[(2r,4s,5s,6s)-5-hydroxy-6-methyl-4-morpholin-4-yloxan-2-yl]oxy-4-methoxy-8,10-dihydro-7h-tetracene-5,12-dione;hydrochloride Chemical compound Cl.N1([C@H]2C[C@@H](O[C@@H](C)[C@H]2O)O[C@H]2C[C@@](O)(CC=3C(O)=C4C(=O)C=5C=CC=C(C=5C(=O)C4=C(O)C=32)OC)C(=O)CO)CCOCC1 INAUWOVKEZHHDM-PEDBPRJASA-N 0.000 description 1
- RCFNNLSZHVHCEK-IMHLAKCZSA-N (7s,9s)-7-(4-amino-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)C1CC([NH3+])CC(C)O1 RCFNNLSZHVHCEK-IMHLAKCZSA-N 0.000 description 1
- NOPNWHSMQOXAEI-PUCKCBAPSA-N (7s,9s)-7-[(2r,4s,5s,6s)-4-(2,3-dihydropyrrol-1-yl)-5-hydroxy-6-methyloxan-2-yl]oxy-6,9,11-trihydroxy-9-(2-hydroxyacetyl)-4-methoxy-8,10-dihydro-7h-tetracene-5,12-dione Chemical compound N1([C@H]2C[C@@H](O[C@@H](C)[C@H]2O)O[C@H]2C[C@@](O)(CC=3C(O)=C4C(=O)C=5C=CC=C(C=5C(=O)C4=C(O)C=32)OC)C(=O)CO)CCC=C1 NOPNWHSMQOXAEI-PUCKCBAPSA-N 0.000 description 1
- FPVKHBSQESCIEP-UHFFFAOYSA-N (8S)-3-(2-deoxy-beta-D-erythro-pentofuranosyl)-3,6,7,8-tetrahydroimidazo[4,5-d][1,3]diazepin-8-ol Natural products C1C(O)C(CO)OC1N1C(NC=NCC2O)=C2N=C1 FPVKHBSQESCIEP-UHFFFAOYSA-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
- AGNGYMCLFWQVGX-AGFFZDDWSA-N (e)-1-[(2s)-2-amino-2-carboxyethoxy]-2-diazonioethenolate Chemical compound OC(=O)[C@@H](N)CO\C([O-])=C\[N+]#N AGNGYMCLFWQVGX-AGFFZDDWSA-N 0.000 description 1
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 1
- 101150084750 1 gene Proteins 0.000 description 1
- FONKWHRXTPJODV-DNQXCXABSA-N 1,3-bis[2-[(8s)-8-(chloromethyl)-4-hydroxy-1-methyl-7,8-dihydro-3h-pyrrolo[3,2-e]indole-6-carbonyl]-1h-indol-5-yl]urea Chemical compound C1([C@H](CCl)CN2C(=O)C=3NC4=CC=C(C=C4C=3)NC(=O)NC=3C=C4C=C(NC4=CC=3)C(=O)N3C4=CC(O)=C5NC=C(C5=C4[C@H](CCl)C3)C)=C2C=C(O)C2=C1C(C)=CN2 FONKWHRXTPJODV-DNQXCXABSA-N 0.000 description 1
- BTOTXLJHDSNXMW-POYBYMJQSA-N 2,3-dideoxyuridine Chemical compound O1[C@H](CO)CC[C@@H]1N1C(=O)NC(=O)C=C1 BTOTXLJHDSNXMW-POYBYMJQSA-N 0.000 description 1
- BOMZMNZEXMAQQW-UHFFFAOYSA-N 2,5,11-trimethyl-6h-pyrido[4,3-b]carbazol-2-ium-9-ol;acetate Chemical compound CC([O-])=O.C[N+]1=CC=C2C(C)=C(NC=3C4=CC(O)=CC=3)C4=C(C)C2=C1 BOMZMNZEXMAQQW-UHFFFAOYSA-N 0.000 description 1
- QCXJFISCRQIYID-IAEPZHFASA-N 2-amino-1-n-[(3s,6s,7r,10s,16s)-3-[(2s)-butan-2-yl]-7,11,14-trimethyl-2,5,9,12,15-pentaoxo-10-propan-2-yl-8-oxa-1,4,11,14-tetrazabicyclo[14.3.0]nonadecan-6-yl]-4,6-dimethyl-3-oxo-9-n-[(3s,6s,7r,10s,16s)-7,11,14-trimethyl-2,5,9,12,15-pentaoxo-3,10-di(propa 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=C2C(C(=O)N[C@@H]3C(=O)N[C@H](C(N4CCC[C@H]4C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]3C)=O)[C@@H](C)CC)=C(N)C(=O)C(C)=C2O2)C2=C(C)C=C1 QCXJFISCRQIYID-IAEPZHFASA-N 0.000 description 1
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 1
- VNBAOSVONFJBKP-UHFFFAOYSA-N 2-chloro-n,n-bis(2-chloroethyl)propan-1-amine;hydrochloride Chemical compound Cl.CC(Cl)CN(CCCl)CCCl VNBAOSVONFJBKP-UHFFFAOYSA-N 0.000 description 1
- YIMDLWDNDGKDTJ-QLKYHASDSA-N 3'-deamino-3'-(3-cyanomorpholin-4-yl)doxorubicin Chemical compound N1([C@H]2C[C@@H](O[C@@H](C)[C@H]2O)O[C@H]2C[C@@](O)(CC=3C(O)=C4C(=O)C=5C=CC=C(C=5C(=O)C4=C(O)C=32)OC)C(=O)CO)CCOCC1C#N YIMDLWDNDGKDTJ-QLKYHASDSA-N 0.000 description 1
- NDMPLJNOPCLANR-UHFFFAOYSA-N 3,4-dihydroxy-15-(4-hydroxy-18-methoxycarbonyl-5,18-seco-ibogamin-18-yl)-16-methoxy-1-methyl-6,7-didehydro-aspidospermidine-3-carboxylic acid methyl ester Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 NDMPLJNOPCLANR-UHFFFAOYSA-N 0.000 description 1
- 238000011455 3D conformal radiation therapy Methods 0.000 description 1
- 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 1
- DODQJNMQWMSYGS-QPLCGJKRSA-N 4-[(z)-1-[4-[2-(dimethylamino)ethoxy]phenyl]-1-phenylbut-1-en-2-yl]phenol Chemical compound C=1C=C(O)C=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 DODQJNMQWMSYGS-QPLCGJKRSA-N 0.000 description 1
- TVZGACDUOSZQKY-LBPRGKRZSA-N 4-aminofolic acid Chemical compound C1=NC2=NC(N)=NC(N)=C2N=C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 TVZGACDUOSZQKY-LBPRGKRZSA-N 0.000 description 1
- NMUSYJAQQFHJEW-KVTDHHQDSA-N 5-azacytidine Chemical compound O=C1N=C(N)N=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 NMUSYJAQQFHJEW-KVTDHHQDSA-N 0.000 description 1
- WYXSYVWAUAUWLD-SHUUEZRQSA-N 6-azauridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=N1 WYXSYVWAUAUWLD-SHUUEZRQSA-N 0.000 description 1
- 229960005538 6-diazo-5-oxo-L-norleucine Drugs 0.000 description 1
- YCWQAMGASJSUIP-YFKPBYRVSA-N 6-diazo-5-oxo-L-norleucine Chemical compound OC(=O)[C@@H](N)CCC(=O)C=[N+]=[N-] YCWQAMGASJSUIP-YFKPBYRVSA-N 0.000 description 1
- UFGQWTWQNIGAEB-UHFFFAOYSA-N 7-chloroquinoline-3-carboxylic acid Chemical compound C1=C(Cl)C=CC2=CC(C(=O)O)=CN=C21 UFGQWTWQNIGAEB-UHFFFAOYSA-N 0.000 description 1
- ZGXJTSGNIOSYLO-UHFFFAOYSA-N 88755TAZ87 Chemical compound NCC(=O)CCC(O)=O ZGXJTSGNIOSYLO-UHFFFAOYSA-N 0.000 description 1
- HDZZVAMISRMYHH-UHFFFAOYSA-N 9beta-Ribofuranosyl-7-deazaadenin Natural products C1=CC=2C(N)=NC=NC=2N1C1OC(CO)C(O)C1O HDZZVAMISRMYHH-UHFFFAOYSA-N 0.000 description 1
- 208000030507 AIDS Diseases 0.000 description 1
- 102000000872 ATM Human genes 0.000 description 1
- 208000010507 Adenocarcinoma of Lung Diseases 0.000 description 1
- CEIZFXOZIQNICU-UHFFFAOYSA-N Alternaria alternata Crofton-weed toxin Natural products CCC(C)C1NC(=O)C(C(C)=O)=C1O CEIZFXOZIQNICU-UHFFFAOYSA-N 0.000 description 1
- 244000303258 Annona diversifolia Species 0.000 description 1
- 235000002198 Annona diversifolia Nutrition 0.000 description 1
- 108020004491 Antisense DNA Proteins 0.000 description 1
- 102000014654 Aromatase Human genes 0.000 description 1
- 108010078554 Aromatase Proteins 0.000 description 1
- 241000228212 Aspergillus Species 0.000 description 1
- 206010003571 Astrocytoma Diseases 0.000 description 1
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 description 1
- 208000003950 B-cell lymphoma Diseases 0.000 description 1
- MLDQJTXFUGDVEO-UHFFFAOYSA-N BAY-43-9006 Chemical compound C1=NC(C(=O)NC)=CC(OC=2C=CC(NC(=O)NC=3C=C(C(Cl)=CC=3)C(F)(F)F)=CC=2)=C1 MLDQJTXFUGDVEO-UHFFFAOYSA-N 0.000 description 1
- 108091007743 BRCA1/2 Proteins 0.000 description 1
- VGGGPCQERPFHOB-MCIONIFRSA-N Bestatin Chemical compound CC(C)C[C@H](C(O)=O)NC(=O)[C@@H](O)[C@H](N)CC1=CC=CC=C1 VGGGPCQERPFHOB-MCIONIFRSA-N 0.000 description 1
- 206010005949 Bone cancer Diseases 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- MBABCNBNDNGODA-LTGLSHGVSA-N Bullatacin Natural products O=C1C(C[C@H](O)CCCCCCCCCC[C@@H](O)[C@@H]2O[C@@H]([C@@H]3O[C@H]([C@@H](O)CCCCCCCCCC)CC3)CC2)=C[C@H](C)O1 MBABCNBNDNGODA-LTGLSHGVSA-N 0.000 description 1
- KGGVWMAPBXIMEM-ZRTAFWODSA-N Bullatacinone Chemical compound O1[C@@H]([C@@H](O)CCCCCCCCCC)CC[C@@H]1[C@@H]1O[C@@H]([C@H](O)CCCCCCCCCC[C@H]2OC(=O)[C@H](CC(C)=O)C2)CC1 KGGVWMAPBXIMEM-ZRTAFWODSA-N 0.000 description 1
- KGGVWMAPBXIMEM-JQFCFGFHSA-N Bullatacinone Natural products O=C(C[C@H]1C(=O)O[C@H](CCCCCCCCCC[C@H](O)[C@@H]2O[C@@H]([C@@H]3O[C@@H]([C@@H](O)CCCCCCCCCC)CC3)CC2)C1)C KGGVWMAPBXIMEM-JQFCFGFHSA-N 0.000 description 1
- OBMZMSLWNNWEJA-XNCRXQDQSA-N C1=CC=2C(C[C@@H]3NC(=O)[C@@H](NC(=O)[C@H](NC(=O)N(CC#CCN(CCCC[C@H](NC(=O)[C@@H](CC4=CC=CC=C4)NC3=O)C(=O)N)CC=C)NC(=O)[C@@H](N)C)CC3=CNC4=C3C=CC=C4)C)=CNC=2C=C1 Chemical compound C1=CC=2C(C[C@@H]3NC(=O)[C@@H](NC(=O)[C@H](NC(=O)N(CC#CCN(CCCC[C@H](NC(=O)[C@@H](CC4=CC=CC=C4)NC3=O)C(=O)N)CC=C)NC(=O)[C@@H](N)C)CC3=CNC4=C3C=CC=C4)C)=CNC=2C=C1 OBMZMSLWNNWEJA-XNCRXQDQSA-N 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- SHHKQEUPHAENFK-UHFFFAOYSA-N Carboquone Chemical compound O=C1C(C)=C(N2CC2)C(=O)C(C(COC(N)=O)OC)=C1N1CC1 SHHKQEUPHAENFK-UHFFFAOYSA-N 0.000 description 1
- 201000009030 Carcinoma Diseases 0.000 description 1
- AOCCBINRVIKJHY-UHFFFAOYSA-N Carmofur Chemical compound CCCCCCNC(=O)N1C=C(F)C(=O)NC1=O AOCCBINRVIKJHY-UHFFFAOYSA-N 0.000 description 1
- 101150053721 Cdk5 gene Proteins 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- JWBOIMRXGHLCPP-UHFFFAOYSA-N Chloditan Chemical compound C=1C=CC=C(Cl)C=1C(C(Cl)Cl)C1=CC=C(Cl)C=C1 JWBOIMRXGHLCPP-UHFFFAOYSA-N 0.000 description 1
- XCDXSSFOJZZGQC-UHFFFAOYSA-N Chlornaphazine Chemical compound C1=CC=CC2=CC(N(CCCl)CCCl)=CC=C21 XCDXSSFOJZZGQC-UHFFFAOYSA-N 0.000 description 1
- MKQWTWSXVILIKJ-LXGUWJNJSA-N Chlorozotocin Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](C=O)NC(=O)N(N=O)CCCl MKQWTWSXVILIKJ-LXGUWJNJSA-N 0.000 description 1
- 108091060290 Chromatid Proteins 0.000 description 1
- 108010077544 Chromatin Proteins 0.000 description 1
- 208000010833 Chronic myeloid leukaemia Diseases 0.000 description 1
- GUTLYIVDDKVIGB-OUBTZVSYSA-N Cobalt-60 Chemical compound [60Co] GUTLYIVDDKVIGB-OUBTZVSYSA-N 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 229930188224 Cryptophycin Natural products 0.000 description 1
- 108020005124 DNA Adducts Proteins 0.000 description 1
- 102000003915 DNA Topoisomerases Human genes 0.000 description 1
- 108090000323 DNA Topoisomerases Proteins 0.000 description 1
- 108010041986 DNA Vaccines Proteins 0.000 description 1
- 229940126161 DNA alkylating agent Drugs 0.000 description 1
- 108091008102 DNA aptamers Proteins 0.000 description 1
- 230000008265 DNA repair mechanism Effects 0.000 description 1
- 229940021995 DNA vaccine Drugs 0.000 description 1
- 241000450599 DNA viruses Species 0.000 description 1
- 230000004568 DNA-binding Effects 0.000 description 1
- 241000252212 Danio rerio Species 0.000 description 1
- WEAHRLBPCANXCN-UHFFFAOYSA-N Daunomycin Natural products CCC1(O)CC(OC2CC(N)C(O)C(C)O2)c3cc4C(=O)c5c(OC)cccc5C(=O)c4c(O)c3C1 WEAHRLBPCANXCN-UHFFFAOYSA-N 0.000 description 1
- NNJPGOLRFBJNIW-UHFFFAOYSA-N Demecolcine Natural products C1=C(OC)C(=O)C=C2C(NC)CCC3=CC(OC)=C(OC)C(OC)=C3C2=C1 NNJPGOLRFBJNIW-UHFFFAOYSA-N 0.000 description 1
- 108010002156 Depsipeptides Proteins 0.000 description 1
- AUGQEEXBDZWUJY-ZLJUKNTDSA-N Diacetoxyscirpenol Chemical compound C([C@]12[C@]3(C)[C@H](OC(C)=O)[C@@H](O)[C@H]1O[C@@H]1C=C(C)CC[C@@]13COC(=O)C)O2 AUGQEEXBDZWUJY-ZLJUKNTDSA-N 0.000 description 1
- AUGQEEXBDZWUJY-UHFFFAOYSA-N Diacetoxyscirpenol Natural products CC(=O)OCC12CCC(C)=CC1OC1C(O)C(OC(C)=O)C2(C)C11CO1 AUGQEEXBDZWUJY-UHFFFAOYSA-N 0.000 description 1
- 241000255581 Drosophila <fruit fly, genus> Species 0.000 description 1
- 101100300807 Drosophila melanogaster spn-A gene Proteins 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- 229930193152 Dynemicin Natural products 0.000 description 1
- AFMYMMXSQGUCBK-UHFFFAOYSA-N Endynamicin A Natural products C1#CC=CC#CC2NC(C=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C(O)=C3)=C3C34OC32C(C)C(C(O)=O)=C(OC)C41 AFMYMMXSQGUCBK-UHFFFAOYSA-N 0.000 description 1
- SAMRUMKYXPVKPA-VFKOLLTISA-N Enocitabine Chemical compound O=C1N=C(NC(=O)CCCCCCCCCCCCCCCCCCCCC)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 SAMRUMKYXPVKPA-VFKOLLTISA-N 0.000 description 1
- 241000991587 Enterovirus C Species 0.000 description 1
- OBMLHUPNRURLOK-XGRAFVIBSA-N Epitiostanol Chemical compound C1[C@@H]2S[C@@H]2C[C@]2(C)[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CC[C@H]21 OBMLHUPNRURLOK-XGRAFVIBSA-N 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 229930189413 Esperamicin Natural products 0.000 description 1
- 108700039887 Essential Genes Proteins 0.000 description 1
- JOYRKODLDBILNP-UHFFFAOYSA-N Ethyl urethane Chemical compound CCOC(N)=O JOYRKODLDBILNP-UHFFFAOYSA-N 0.000 description 1
- 241001316028 Euphaedusa tau Species 0.000 description 1
- 241000282324 Felis Species 0.000 description 1
- 101000941893 Felis catus Leucine-rich repeat and calponin homology domain-containing protein 1 Proteins 0.000 description 1
- 241000589601 Francisella Species 0.000 description 1
- 208000002339 Frontotemporal Lobar Degeneration Diseases 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 230000010337 G2 phase Effects 0.000 description 1
- 208000034951 Genetic Translocation Diseases 0.000 description 1
- 208000031448 Genomic Instability Diseases 0.000 description 1
- 208000032320 Germ cell tumor of testis Diseases 0.000 description 1
- 102000019058 Glycogen Synthase Kinase 3 beta Human genes 0.000 description 1
- 108010051975 Glycogen Synthase Kinase 3 beta Proteins 0.000 description 1
- BLCLNMBMMGCOAS-URPVMXJPSA-N Goserelin Chemical compound C([C@@H](C(=O)N[C@H](COC(C)(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCN=C(N)N)C(=O)N1[C@@H](CCC1)C(=O)NNC(N)=O)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H]1NC(=O)CC1)C1=CC=C(O)C=C1 BLCLNMBMMGCOAS-URPVMXJPSA-N 0.000 description 1
- 108010069236 Goserelin Proteins 0.000 description 1
- 108020005004 Guide RNA Proteins 0.000 description 1
- 229940121710 HMGCoA reductase inhibitor Drugs 0.000 description 1
- 241000713858 Harvey murine sarcoma virus Species 0.000 description 1
- 102100034051 Heat shock protein HSP 90-alpha Human genes 0.000 description 1
- 108010033040 Histones Proteins 0.000 description 1
- 208000017604 Hodgkin disease Diseases 0.000 description 1
- 208000021519 Hodgkin lymphoma Diseases 0.000 description 1
- 208000010747 Hodgkins lymphoma Diseases 0.000 description 1
- 101001016865 Homo sapiens Heat shock protein HSP 90-alpha Proteins 0.000 description 1
- 101000581326 Homo sapiens Mediator of DNA damage checkpoint protein 1 Proteins 0.000 description 1
- 101000616438 Homo sapiens Microtubule-associated protein 4 Proteins 0.000 description 1
- 101000777293 Homo sapiens Serine/threonine-protein kinase Chk1 Proteins 0.000 description 1
- 101000777277 Homo sapiens Serine/threonine-protein kinase Chk2 Proteins 0.000 description 1
- 101001059443 Homo sapiens Serine/threonine-protein kinase MARK1 Proteins 0.000 description 1
- 101000891649 Homo sapiens Transcription elongation factor A protein-like 1 Proteins 0.000 description 1
- 206010062904 Hormone-refractory prostate cancer Diseases 0.000 description 1
- 241000701109 Human adenovirus 2 Species 0.000 description 1
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 1
- VSNHCAURESNICA-UHFFFAOYSA-N Hydroxyurea Chemical compound NC(=O)NO VSNHCAURESNICA-UHFFFAOYSA-N 0.000 description 1
- 206010020843 Hyperthermia Diseases 0.000 description 1
- MPBVHIBUJCELCL-UHFFFAOYSA-N Ibandronate Chemical compound CCCCCN(C)CCC(O)(P(O)(O)=O)P(O)(O)=O MPBVHIBUJCELCL-UHFFFAOYSA-N 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 102000009786 Immunoglobulin Constant Regions Human genes 0.000 description 1
- 108010009817 Immunoglobulin Constant Regions Proteins 0.000 description 1
- 102000012214 Immunoproteins Human genes 0.000 description 1
- 108010036650 Immunoproteins Proteins 0.000 description 1
- 206010061252 Intraocular melanoma Diseases 0.000 description 1
- 208000007766 Kaposi sarcoma Diseases 0.000 description 1
- YQEZLKZALYSWHR-UHFFFAOYSA-N Ketamine Chemical compound C=1C=CC=C(Cl)C=1C1(NC)CCCCC1=O YQEZLKZALYSWHR-UHFFFAOYSA-N 0.000 description 1
- 241000235649 Kluyveromyces Species 0.000 description 1
- 239000002147 L01XE04 - Sunitinib Substances 0.000 description 1
- 239000005511 L01XE05 - Sorafenib Substances 0.000 description 1
- JLERVPBPJHKRBJ-UHFFFAOYSA-N LY 117018 Chemical compound C1=CC(O)=CC=C1C1=C(C(=O)C=2C=CC(OCCN3CCCC3)=CC=2)C2=CC=C(O)C=C2S1 JLERVPBPJHKRBJ-UHFFFAOYSA-N 0.000 description 1
- 208000031671 Large B-Cell Diffuse Lymphoma Diseases 0.000 description 1
- 229920001491 Lentinan Polymers 0.000 description 1
- 241000713666 Lentivirus Species 0.000 description 1
- 108010000817 Leuprolide Proteins 0.000 description 1
- 102000003960 Ligases Human genes 0.000 description 1
- 108090000364 Ligases Proteins 0.000 description 1
- 239000000232 Lipid Bilayer Substances 0.000 description 1
- 208000031422 Lymphocytic Chronic B-Cell Leukemia Diseases 0.000 description 1
- 102000043136 MAP kinase family Human genes 0.000 description 1
- 108091054455 MAP kinase family Proteins 0.000 description 1
- 206010025557 Malignant fibrous histiocytoma of bone Diseases 0.000 description 1
- VJRAUFKOOPNFIQ-UHFFFAOYSA-N Marcellomycin Natural products C12=C(O)C=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C=C2C(C(=O)OC)C(CC)(O)CC1OC(OC1C)CC(N(C)C)C1OC(OC1C)CC(O)C1OC1CC(O)C(O)C(C)O1 VJRAUFKOOPNFIQ-UHFFFAOYSA-N 0.000 description 1
- 229930126263 Maytansine Natural products 0.000 description 1
- 102100027643 Mediator of DNA damage checkpoint protein 1 Human genes 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- IVDYZAAPOLNZKG-KWHRADDSSA-N Mepitiostane Chemical compound O([C@@H]1[C@]2(CC[C@@H]3[C@@]4(C)C[C@H]5S[C@H]5C[C@@H]4CC[C@H]3[C@@H]2CC1)C)C1(OC)CCCC1 IVDYZAAPOLNZKG-KWHRADDSSA-N 0.000 description 1
- 102100021794 Microtubule-associated protein 4 Human genes 0.000 description 1
- 102000003979 Mineralocorticoid Receptors Human genes 0.000 description 1
- 108090000375 Mineralocorticoid Receptors Proteins 0.000 description 1
- 229930192392 Mitomycin Natural products 0.000 description 1
- 241000713869 Moloney murine leukemia virus Species 0.000 description 1
- 102000006833 Multifunctional Enzymes Human genes 0.000 description 1
- 108010047290 Multifunctional Enzymes Proteins 0.000 description 1
- 208000034578 Multiple myelomas Diseases 0.000 description 1
- 241000204795 Muraena helena Species 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 101000596402 Mus musculus Neuronal vesicle trafficking-associated protein 1 Proteins 0.000 description 1
- 101100365003 Mus musculus Scel gene Proteins 0.000 description 1
- 101000800539 Mus musculus Translationally-controlled tumor protein Proteins 0.000 description 1
- OVBPIULPVIDEAO-UHFFFAOYSA-N N-Pteroyl-L-glutaminsaeure Natural products C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)NC(CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-UHFFFAOYSA-N 0.000 description 1
- SYNHCENRCUAUNM-UHFFFAOYSA-N Nitrogen mustard N-oxide hydrochloride Chemical compound Cl.ClCC[N+]([O-])(C)CCCl SYNHCENRCUAUNM-UHFFFAOYSA-N 0.000 description 1
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 description 1
- 102000007999 Nuclear Proteins Human genes 0.000 description 1
- 108010089610 Nuclear Proteins Proteins 0.000 description 1
- 206010030155 Oesophageal carcinoma Diseases 0.000 description 1
- 108010038807 Oligopeptides Proteins 0.000 description 1
- 102000015636 Oligopeptides Human genes 0.000 description 1
- 229930187135 Olivomycin Natural products 0.000 description 1
- 108700020796 Oncogene Proteins 0.000 description 1
- 102000043276 Oncogene Human genes 0.000 description 1
- VREZDOWOLGNDPW-ALTGWBOUSA-N Pancratistatin Chemical compound C1=C2[C@H]3[C@@H](O)[C@H](O)[C@@H](O)[C@@H](O)[C@@H]3NC(=O)C2=C(O)C2=C1OCO2 VREZDOWOLGNDPW-ALTGWBOUSA-N 0.000 description 1
- VREZDOWOLGNDPW-MYVCAWNPSA-N Pancratistatin Natural products O=C1N[C@H]2[C@H](O)[C@H](O)[C@H](O)[C@H](O)[C@@H]2c2c1c(O)c1OCOc1c2 VREZDOWOLGNDPW-MYVCAWNPSA-N 0.000 description 1
- 108090000526 Papain Proteins 0.000 description 1
- 241001631646 Papillomaviridae Species 0.000 description 1
- 229930040373 Paraformaldehyde Natural products 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 108010057150 Peplomycin Proteins 0.000 description 1
- 101710176384 Peptide 1 Proteins 0.000 description 1
- 108010079855 Peptide Aptamers Proteins 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 208000000609 Pick Disease of the Brain Diseases 0.000 description 1
- KMSKQZKKOZQFFG-HSUXVGOQSA-N Pirarubicin Chemical compound O([C@H]1[C@@H](N)C[C@@H](O[C@H]1C)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]1CCCCO1 KMSKQZKKOZQFFG-HSUXVGOQSA-N 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 241000276498 Pollachius virens Species 0.000 description 1
- 241001505332 Polyomavirus sp. Species 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 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
- 208000010291 Primary Progressive Nonfluent Aphasia Diseases 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 102000004245 Proteasome Endopeptidase Complex Human genes 0.000 description 1
- 108090000708 Proteasome Endopeptidase Complex Proteins 0.000 description 1
- 239000012083 RIPA buffer Substances 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- AHHFEZNOXOZZQA-ZEBDFXRSSA-N Ranimustine Chemical compound CO[C@H]1O[C@H](CNC(=O)N(CCCl)N=O)[C@@H](O)[C@H](O)[C@H]1O AHHFEZNOXOZZQA-ZEBDFXRSSA-N 0.000 description 1
- 201000000582 Retinoblastoma Diseases 0.000 description 1
- 206010038997 Retroviral infections 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
- 102000006382 Ribonucleases Human genes 0.000 description 1
- 108010083644 Ribonucleases Proteins 0.000 description 1
- 108091028664 Ribonucleotide Proteins 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 230000018199 S phase Effects 0.000 description 1
- 241000235070 Saccharomyces Species 0.000 description 1
- 101000781972 Schizosaccharomyces pombe (strain 972 / ATCC 24843) Protein wos2 Proteins 0.000 description 1
- 201000010208 Seminoma Diseases 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 102100031081 Serine/threonine-protein kinase Chk1 Human genes 0.000 description 1
- 102100031075 Serine/threonine-protein kinase Chk2 Human genes 0.000 description 1
- 229920000519 Sizofiran Polymers 0.000 description 1
- KEAYESYHFKHZAL-UHFFFAOYSA-N Sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 1
- 208000000102 Squamous Cell Carcinoma of Head and Neck Diseases 0.000 description 1
- 108010090804 Streptavidin Proteins 0.000 description 1
- 241000193996 Streptococcus pyogenes Species 0.000 description 1
- 206010042566 Superinfection Diseases 0.000 description 1
- BXFOFFBJRFZBQZ-QYWOHJEZSA-N T-2 toxin Chemical compound C([C@@]12[C@]3(C)[C@H](OC(C)=O)[C@@H](O)[C@H]1O[C@H]1[C@]3(COC(C)=O)C[C@@H](C(=C1)C)OC(=O)CC(C)C)O2 BXFOFFBJRFZBQZ-QYWOHJEZSA-N 0.000 description 1
- 208000034799 Tauopathies Diseases 0.000 description 1
- FRJSECSOXKQMOD-HQRMLTQVSA-N Taxa-4(5),11(12)-diene Chemical group C1C[C@]2(C)CCC=C(C)[C@H]2C[C@@H]2CCC(C)=C1C2(C)C FRJSECSOXKQMOD-HQRMLTQVSA-N 0.000 description 1
- 241001116500 Taxus Species 0.000 description 1
- BPEGJWRSRHCHSN-UHFFFAOYSA-N Temozolomide Chemical compound O=C1N(C)N=NC2=C(C(N)=O)N=CN21 BPEGJWRSRHCHSN-UHFFFAOYSA-N 0.000 description 1
- CGMTUJFWROPELF-UHFFFAOYSA-N Tenuazonic acid Natural products CCC(C)C1NC(=O)C(=C(C)/O)C1=O CGMTUJFWROPELF-UHFFFAOYSA-N 0.000 description 1
- 208000024313 Testicular Neoplasms Diseases 0.000 description 1
- 206010057644 Testis cancer Diseases 0.000 description 1
- 102100036407 Thioredoxin Human genes 0.000 description 1
- IWEQQRMGNVVKQW-OQKDUQJOSA-N Toremifene citrate Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O.C1=CC(OCCN(C)C)=CC=C1C(\C=1C=CC=CC=1)=C(\CCCl)C1=CC=CC=C1 IWEQQRMGNVVKQW-OQKDUQJOSA-N 0.000 description 1
- 101001009610 Toxoplasma gondii Dense granule protein 5 Proteins 0.000 description 1
- 102100040250 Transcription elongation factor A protein-like 1 Human genes 0.000 description 1
- 241000223259 Trichoderma Species 0.000 description 1
- UMILHIMHKXVDGH-UHFFFAOYSA-N Triethylene glycol diglycidyl ether Chemical compound C1OC1COCCOCCOCCOCC1CO1 UMILHIMHKXVDGH-UHFFFAOYSA-N 0.000 description 1
- 102000044209 Tumor Suppressor Genes Human genes 0.000 description 1
- 108700025716 Tumor Suppressor Genes Proteins 0.000 description 1
- 241000288668 Tupaiidae Species 0.000 description 1
- 102000007537 Type II DNA Topoisomerases Human genes 0.000 description 1
- 108010046308 Type II DNA Topoisomerases Proteins 0.000 description 1
- 208000015778 Undifferentiated pleomorphic sarcoma Diseases 0.000 description 1
- 229910052770 Uranium Inorganic materials 0.000 description 1
- 208000002495 Uterine Neoplasms Diseases 0.000 description 1
- 201000005969 Uveal melanoma Diseases 0.000 description 1
- 241000700618 Vaccinia virus Species 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 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
- 108020000999 Viral RNA Proteins 0.000 description 1
- 102100036973 X-ray repair cross-complementing protein 5 Human genes 0.000 description 1
- 101710124921 X-ray repair cross-complementing protein 5 Proteins 0.000 description 1
- 102100036976 X-ray repair cross-complementing protein 6 Human genes 0.000 description 1
- 101710124907 X-ray repair cross-complementing protein 6 Proteins 0.000 description 1
- 241000269457 Xenopus tropicalis Species 0.000 description 1
- ZYVSOIYQKUDENJ-ASUJBHBQSA-N [(2R,3R,4R,6R)-6-[[(6S,7S)-6-[(2S,4R,5R,6R)-4-[(2R,4R,5R,6R)-4-[(2S,4S,5S,6S)-5-acetyloxy-4-hydroxy-4,6-dimethyloxan-2-yl]oxy-5-hydroxy-6-methyloxan-2-yl]oxy-5-hydroxy-6-methyloxan-2-yl]oxy-7-[(3S,4R)-3,4-dihydroxy-1-methoxy-2-oxopentyl]-4,10-dihydroxy-3-methyl-5-oxo-7,8-dihydro-6H-anthracen-2-yl]oxy]-4-[(2R,4R,5R,6R)-4-hydroxy-5-methoxy-6-methyloxan-2-yl]oxy-2-methyloxan-3-yl] acetate Chemical class COC([C@@H]1Cc2cc3cc(O[C@@H]4C[C@@H](O[C@@H]5C[C@@H](O)[C@@H](OC)[C@@H](C)O5)[C@H](OC(C)=O)[C@@H](C)O4)c(C)c(O)c3c(O)c2C(=O)[C@H]1O[C@H]1C[C@@H](O[C@@H]2C[C@@H](O[C@H]3C[C@](C)(O)[C@@H](OC(C)=O)[C@H](C)O3)[C@H](O)[C@@H](C)O2)[C@H](O)[C@@H](C)O1)C(=O)[C@@H](O)[C@@H](C)O ZYVSOIYQKUDENJ-ASUJBHBQSA-N 0.000 description 1
- UKFDBDGJFYRBHE-FKGSWUQWSA-N [(3e,6s,7s,9s)-5-[(2r,3s)-3-acetyloxy-2-(2-hydroxyethyl)oxetan-3-yl]-3,7-dihydroxy-9-[(2r,3s)-2-hydroxy-5-methyl-3-[(2-methylpropan-2-yl)oxycarbonylamino]hexanoyl]oxy-4,10,11,11-tetramethyl-2-oxo-6-bicyclo[5.3.1]undeca-1(10),3-dienyl] benzoate Chemical compound O([C@@H]1[C@]2(O)C[C@@H](C(=C(C(=O)\C(O)=C(C)/C1[C@]1([C@H](OC1)CCO)OC(C)=O)C2(C)C)C)OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)CC(C)C)C(=O)C1=CC=CC=C1 UKFDBDGJFYRBHE-FKGSWUQWSA-N 0.000 description 1
- SPJCRMJCFSJKDE-ZWBUGVOYSA-N [(3s,8s,9s,10r,13r,14s,17r)-10,13-dimethyl-17-[(2r)-6-methylheptan-2-yl]-2,3,4,7,8,9,11,12,14,15,16,17-dodecahydro-1h-cyclopenta[a]phenanthren-3-yl] 2-[4-[bis(2-chloroethyl)amino]phenyl]acetate Chemical compound O([C@@H]1CC2=CC[C@H]3[C@@H]4CC[C@@H]([C@]4(CC[C@@H]3[C@@]2(C)CC1)C)[C@H](C)CCCC(C)C)C(=O)CC1=CC=C(N(CCCl)CCCl)C=C1 SPJCRMJCFSJKDE-ZWBUGVOYSA-N 0.000 description 1
- IFJUINDAXYAPTO-UUBSBJJBSA-N [(8r,9s,13s,14s,17s)-17-[2-[4-[4-[bis(2-chloroethyl)amino]phenyl]butanoyloxy]acetyl]oxy-13-methyl-6,7,8,9,11,12,14,15,16,17-decahydrocyclopenta[a]phenanthren-3-yl] benzoate Chemical compound C([C@@H]1[C@@H](C2=CC=3)CC[C@]4([C@H]1CC[C@@H]4OC(=O)COC(=O)CCCC=1C=CC(=CC=1)N(CCCl)CCCl)C)CC2=CC=3OC(=O)C1=CC=CC=C1 IFJUINDAXYAPTO-UUBSBJJBSA-N 0.000 description 1
- IHGLINDYFMDHJG-UHFFFAOYSA-N [2-(4-methoxyphenyl)-3,4-dihydronaphthalen-1-yl]-[4-(2-pyrrolidin-1-ylethoxy)phenyl]methanone Chemical compound C1=CC(OC)=CC=C1C(CCC1=CC=CC=C11)=C1C(=O)C(C=C1)=CC=C1OCCN1CCCC1 IHGLINDYFMDHJG-UHFFFAOYSA-N 0.000 description 1
- XZSRRNFBEIOBDA-CFNBKWCHSA-N [2-[(2s,4s)-4-[(2r,4s,5s,6s)-4-amino-5-hydroxy-6-methyloxan-2-yl]oxy-2,5,12-trihydroxy-7-methoxy-6,11-dioxo-3,4-dihydro-1h-tetracen-2-yl]-2-oxoethyl] 2,2-diethoxyacetate Chemical compound O([C@H]1C[C@](CC2=C(O)C=3C(=O)C4=CC=CC(OC)=C4C(=O)C=3C(O)=C21)(O)C(=O)COC(=O)C(OCC)OCC)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 XZSRRNFBEIOBDA-CFNBKWCHSA-N 0.000 description 1
- 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 1
- ZOZKYEHVNDEUCO-XUTVFYLZSA-N aceglatone Chemical compound O1C(=O)[C@H](OC(C)=O)[C@@H]2OC(=O)[C@@H](OC(=O)C)[C@@H]21 ZOZKYEHVNDEUCO-XUTVFYLZSA-N 0.000 description 1
- 229950002684 aceglatone Drugs 0.000 description 1
- 229930183665 actinomycin Natural products 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
- 230000001154 acute effect Effects 0.000 description 1
- 210000005006 adaptive immune system Anatomy 0.000 description 1
- 102000035181 adaptor proteins Human genes 0.000 description 1
- 108091005764 adaptor proteins Proteins 0.000 description 1
- 239000002487 adenosine deaminase inhibitor Substances 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 229950004955 adozelesin Drugs 0.000 description 1
- BYRVKDUQDLJUBX-JJCDCTGGSA-N adozelesin Chemical compound C1=CC=C2OC(C(=O)NC=3C=C4C=C(NC4=CC=3)C(=O)N3C[C@H]4C[C@]44C5=C(C(C=C43)=O)NC=C5C)=CC2=C1 BYRVKDUQDLJUBX-JJCDCTGGSA-N 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
- 229960000473 altretamine Drugs 0.000 description 1
- 229960003437 aminoglutethimide Drugs 0.000 description 1
- ROBVIMPUHSLWNV-UHFFFAOYSA-N aminoglutethimide Chemical compound C=1C=C(N)C=CC=1C1(CC)CCC(=O)NC1=O ROBVIMPUHSLWNV-UHFFFAOYSA-N 0.000 description 1
- 229960002749 aminolevulinic acid Drugs 0.000 description 1
- 229960003896 aminopterin Drugs 0.000 description 1
- BBDAGFIXKZCXAH-CCXZUQQUSA-N ancitabine Chemical compound N=C1C=CN2[C@@H]3O[C@H](CO)[C@@H](O)[C@@H]3OC2=N1 BBDAGFIXKZCXAH-CCXZUQQUSA-N 0.000 description 1
- 229950000242 ancitabine Drugs 0.000 description 1
- 239000003098 androgen Substances 0.000 description 1
- 229940030486 androgens Drugs 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 229940045799 anthracyclines and related substance Drugs 0.000 description 1
- 230000002280 anti-androgenic effect Effects 0.000 description 1
- 229940046836 anti-estrogen Drugs 0.000 description 1
- 230000001833 anti-estrogenic effect Effects 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 239000000051 antiandrogen Substances 0.000 description 1
- 229940030495 antiandrogen sex hormone and modulator of the genital system Drugs 0.000 description 1
- 230000005875 antibody response Effects 0.000 description 1
- 239000013059 antihormonal agent Substances 0.000 description 1
- 229940100197 antimetabolite Drugs 0.000 description 1
- 229940045687 antimetabolites folic acid analogs Drugs 0.000 description 1
- 229940045719 antineoplastic alkylating agent nitrosoureas Drugs 0.000 description 1
- 239000003816 antisense DNA Substances 0.000 description 1
- 150000008209 arabinosides Chemical class 0.000 description 1
- 229960002756 azacitidine Drugs 0.000 description 1
- 229950011321 azaserine Drugs 0.000 description 1
- 150000001541 aziridines Chemical class 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 229960000997 bicalutamide Drugs 0.000 description 1
- 210000003445 biliary tract Anatomy 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 239000003124 biologic agent Substances 0.000 description 1
- 230000031018 biological processes and functions Effects 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 229950008548 bisantrene Drugs 0.000 description 1
- 229950006844 bizelesin Drugs 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- UBJAHGAUPNGZFF-XOVTVWCYSA-N bms-184476 Chemical compound O([C@H]1[C@@H]2[C@]3(OC(C)=O)CO[C@@H]3C[C@@H]([C@]2(C(=O)[C@H](OC(C)=O)C2=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)C=3C=CC=CC=3)C=3C=CC=CC=3)C[C@]1(O)C2(C)C)C)OCSC)C(=O)C1=CC=CC=C1 UBJAHGAUPNGZFF-XOVTVWCYSA-N 0.000 description 1
- GMJWGJSDPOAZTP-MIDYMNAOSA-N bms-188797 Chemical compound O([C@H]1[C@H]2[C@@](C([C@H](OC(C)=O)C3=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)C=4C=CC=CC=4)C=4C=CC=CC=4)C[C@]1(O)C3(C)C)=O)(C)[C@@H](O)C[C@H]1OC[C@]12OC(=O)OC)C(=O)C1=CC=CC=C1 GMJWGJSDPOAZTP-MIDYMNAOSA-N 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- 229940098773 bovine serum albumin Drugs 0.000 description 1
- 229960005520 bryostatin Drugs 0.000 description 1
- MJQUEDHRCUIRLF-TVIXENOKSA-N bryostatin 1 Chemical compound C([C@@H]1CC(/[C@@H]([C@@](C(C)(C)/C=C/2)(O)O1)OC(=O)/C=C/C=C/CCC)=C\C(=O)OC)[C@H]([C@@H](C)O)OC(=O)C[C@H](O)C[C@@H](O1)C[C@H](OC(C)=O)C(C)(C)[C@]1(O)C[C@@H]1C\C(=C\C(=O)OC)C[C@H]\2O1 MJQUEDHRCUIRLF-TVIXENOKSA-N 0.000 description 1
- MUIWQCKLQMOUAT-AKUNNTHJSA-N bryostatin 20 Natural products COC(=O)C=C1C[C@@]2(C)C[C@]3(O)O[C@](C)(C[C@@H](O)CC(=O)O[C@](C)(C[C@@]4(C)O[C@](O)(CC5=CC(=O)O[C@]45C)C(C)(C)C=C[C@@](C)(C1)O2)[C@@H](C)O)C[C@H](OC(=O)C(C)(C)C)C3(C)C MUIWQCKLQMOUAT-AKUNNTHJSA-N 0.000 description 1
- MBABCNBNDNGODA-LUVUIASKSA-N bullatacin Chemical compound O1[C@@H]([C@@H](O)CCCCCCCCCC)CC[C@@H]1[C@@H]1O[C@@H]([C@H](O)CCCCCCCCCC[C@@H](O)CC=2C(O[C@@H](C)C=2)=O)CC1 MBABCNBNDNGODA-LUVUIASKSA-N 0.000 description 1
- 108700002839 cactinomycin Proteins 0.000 description 1
- 229950009908 cactinomycin Drugs 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- IVFYLRMMHVYGJH-PVPPCFLZSA-N calusterone Chemical compound C1C[C@]2(C)[C@](O)(C)CC[C@H]2[C@@H]2[C@@H](C)CC3=CC(=O)CC[C@]3(C)[C@H]21 IVFYLRMMHVYGJH-PVPPCFLZSA-N 0.000 description 1
- 229950009823 calusterone Drugs 0.000 description 1
- 229960002115 carboquone Drugs 0.000 description 1
- 229960003261 carmofur Drugs 0.000 description 1
- 229950007509 carzelesin Drugs 0.000 description 1
- BBZDXMBRAFTCAA-AREMUKBSSA-N carzelesin Chemical compound C1=2NC=C(C)C=2C([C@H](CCl)CN2C(=O)C=3NC4=CC=C(C=C4C=3)NC(=O)C3=CC4=CC=C(C=C4O3)N(CC)CC)=C2C=C1OC(=O)NC1=CC=CC=C1 BBZDXMBRAFTCAA-AREMUKBSSA-N 0.000 description 1
- 108010047060 carzinophilin Proteins 0.000 description 1
- 230000006652 catabolic pathway Effects 0.000 description 1
- 230000012820 cell cycle checkpoint Effects 0.000 description 1
- 230000018486 cell cycle phase Effects 0.000 description 1
- 230000032823 cell division Effects 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 210000003855 cell nucleus Anatomy 0.000 description 1
- 230000007969 cellular immunity Effects 0.000 description 1
- 230000007248 cellular mechanism Effects 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 230000035572 chemosensitivity Effects 0.000 description 1
- 229940044683 chemotherapy drug Drugs 0.000 description 1
- 229950008249 chlornaphazine Drugs 0.000 description 1
- 229960001480 chlorozotocin Drugs 0.000 description 1
- 210000004756 chromatid Anatomy 0.000 description 1
- 210000003483 chromatin Anatomy 0.000 description 1
- 108091006090 chromatin-associated proteins Proteins 0.000 description 1
- 208000032852 chronic lymphocytic leukemia Diseases 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 210000001072 colon Anatomy 0.000 description 1
- 208000029742 colonic neoplasm Diseases 0.000 description 1
- 238000012875 competitive assay Methods 0.000 description 1
- 238000011340 continuous therapy Methods 0.000 description 1
- 238000002711 conventional external beam radiation therapy Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 108010089438 cryptophycin 1 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
- 108010090203 cryptophycin 8 Proteins 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
- 208000035250 cutaneous malignant susceptibility to 1 melanoma Diseases 0.000 description 1
- 238000005520 cutting process Methods 0.000 description 1
- 230000000120 cytopathologic effect Effects 0.000 description 1
- 210000004292 cytoskeleton Anatomy 0.000 description 1
- 229960000640 dactinomycin Drugs 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 229960005052 demecolcine Drugs 0.000 description 1
- 239000000412 dendrimer Substances 0.000 description 1
- 229920000736 dendritic polymer Polymers 0.000 description 1
- 238000000326 densiometry Methods 0.000 description 1
- 229950003913 detorubicin Drugs 0.000 description 1
- 230000001627 detrimental effect Effects 0.000 description 1
- WVYXNIXAMZOZFK-UHFFFAOYSA-N diaziquone Chemical compound O=C1C(NC(=O)OCC)=C(N2CC2)C(=O)C(NC(=O)OCC)=C1N1CC1 WVYXNIXAMZOZFK-UHFFFAOYSA-N 0.000 description 1
- 229950002389 diaziquone Drugs 0.000 description 1
- BNIILDVGGAEEIG-UHFFFAOYSA-L disodium hydrogen phosphate Chemical compound [Na+].[Na+].OP([O-])([O-])=O BNIILDVGGAEEIG-UHFFFAOYSA-L 0.000 description 1
- 229910000397 disodium phosphate Inorganic materials 0.000 description 1
- 235000019800 disodium phosphate Nutrition 0.000 description 1
- 239000002612 dispersion medium Substances 0.000 description 1
- 238000006073 displacement reaction Methods 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 125000000567 diterpene group Chemical group 0.000 description 1
- 239000003534 dna topoisomerase inhibitor Substances 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
- 239000002552 dosage form Substances 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- ZWAOHEXOSAUJHY-ZIYNGMLESA-N doxifluridine Chemical compound O[C@@H]1[C@H](O)[C@@H](C)O[C@H]1N1C(=O)NC(=O)C(F)=C1 ZWAOHEXOSAUJHY-ZIYNGMLESA-N 0.000 description 1
- 229950005454 doxifluridine Drugs 0.000 description 1
- NOTIQUSPUUHHEH-UXOVVSIBSA-N dromostanolone propionate Chemical compound C([C@@H]1CC2)C(=O)[C@H](C)C[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H](OC(=O)CC)[C@@]2(C)CC1 NOTIQUSPUUHHEH-UXOVVSIBSA-N 0.000 description 1
- 239000006196 drop Substances 0.000 description 1
- 229950004683 drostanolone propionate Drugs 0.000 description 1
- 241001493065 dsRNA viruses Species 0.000 description 1
- 229960005501 duocarmycin Drugs 0.000 description 1
- VQNATVDKACXKTF-XELLLNAOSA-N duocarmycin Chemical compound COC1=C(OC)C(OC)=C2NC(C(=O)N3C4=CC(=O)C5=C([C@@]64C[C@@H]6C3)C=C(N5)C(=O)OC)=CC2=C1 VQNATVDKACXKTF-XELLLNAOSA-N 0.000 description 1
- 229930184221 duocarmycin Natural products 0.000 description 1
- AFMYMMXSQGUCBK-AKMKHHNQSA-N dynemicin a Chemical compound C1#C\C=C/C#C[C@@H]2NC(C=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C(O)=C3)=C3[C@@]34O[C@]32[C@@H](C)C(C(O)=O)=C(OC)[C@H]41 AFMYMMXSQGUCBK-AKMKHHNQSA-N 0.000 description 1
- 230000008482 dysregulation Effects 0.000 description 1
- 229960002759 eflornithine Drugs 0.000 description 1
- XOPYFXBZMVTEJF-PDACKIITSA-N eleutherobin Chemical compound C(/[C@H]1[C@H](C(=CC[C@@H]1C(C)C)C)C[C@@H]([C@@]1(C)O[C@@]2(C=C1)OC)OC(=O)\C=C\C=1N=CN(C)C=1)=C2\CO[C@@H]1OC[C@@H](O)[C@@H](O)[C@@H]1OC(C)=O XOPYFXBZMVTEJF-PDACKIITSA-N 0.000 description 1
- XOPYFXBZMVTEJF-UHFFFAOYSA-N eleutherobin Natural products C1=CC2(OC)OC1(C)C(OC(=O)C=CC=1N=CN(C)C=1)CC(C(=CCC1C(C)C)C)C1C=C2COC1OCC(O)C(O)C1OC(C)=O XOPYFXBZMVTEJF-UHFFFAOYSA-N 0.000 description 1
- 229950000549 elliptinium acetate Drugs 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 229950011487 enocitabine Drugs 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 210000002615 epidermis Anatomy 0.000 description 1
- 230000004049 epigenetic modification Effects 0.000 description 1
- 229950002973 epitiostanol Drugs 0.000 description 1
- 229930013356 epothilone Natural products 0.000 description 1
- 150000003883 epothilone derivatives Chemical class 0.000 description 1
- 229950002017 esorubicin Drugs 0.000 description 1
- ITSGNOIFAJAQHJ-BMFNZSJVSA-N esorubicin 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(=O)CO)[C@H]1C[C@H](N)C[C@H](C)O1 ITSGNOIFAJAQHJ-BMFNZSJVSA-N 0.000 description 1
- LJQQFQHBKUKHIS-WJHRIEJJSA-N esperamicin Chemical compound O1CC(NC(C)C)C(OC)CC1OC1C(O)C(NOC2OC(C)C(SC)C(O)C2)C(C)OC1OC1C(\C2=C/CSSSC)=C(NC(=O)OC)C(=O)C(OC3OC(C)C(O)C(OC(=O)C=4C(=CC(OC)=C(OC)C=4)NC(=O)C(=C)OC)C3)C2(O)C#C\C=C/C#C1 LJQQFQHBKUKHIS-WJHRIEJJSA-N 0.000 description 1
- 239000000328 estrogen antagonist Substances 0.000 description 1
- QSRLNKCNOLVZIR-KRWDZBQOSA-N ethyl (2s)-2-[[2-[4-[bis(2-chloroethyl)amino]phenyl]acetyl]amino]-4-methylsulfanylbutanoate Chemical compound CCOC(=O)[C@H](CCSC)NC(=O)CC1=CC=C(N(CCCl)CCCl)C=C1 QSRLNKCNOLVZIR-KRWDZBQOSA-N 0.000 description 1
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- 229960005237 etoglucid Drugs 0.000 description 1
- 208000021045 exocrine pancreatic carcinoma Diseases 0.000 description 1
- 239000013604 expression vector Substances 0.000 description 1
- 238000002710 external beam radiation therapy Methods 0.000 description 1
- 229940043168 fareston Drugs 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 229960000390 fludarabine Drugs 0.000 description 1
- 229960005304 fludarabine phosphate Drugs 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 229960002074 flutamide 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
- 229960000304 folic acid Drugs 0.000 description 1
- 235000019152 folic acid Nutrition 0.000 description 1
- 239000011724 folic acid Substances 0.000 description 1
- 239000004052 folic acid antagonist Substances 0.000 description 1
- 150000002224 folic acids Chemical class 0.000 description 1
- 230000003325 follicular Effects 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 210000000232 gallbladder Anatomy 0.000 description 1
- 229940044658 gallium nitrate Drugs 0.000 description 1
- 238000012226 gene silencing method Methods 0.000 description 1
- 238000001415 gene therapy Methods 0.000 description 1
- 230000004077 genetic alteration Effects 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 238000010362 genome editing Methods 0.000 description 1
- 229930182470 glycoside Natural products 0.000 description 1
- 150000002338 glycosides Chemical class 0.000 description 1
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 1
- 239000010931 gold Substances 0.000 description 1
- 229910052737 gold Inorganic materials 0.000 description 1
- 229960002913 goserelin Drugs 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 210000004524 haematopoietic cell Anatomy 0.000 description 1
- 230000009931 harmful effect Effects 0.000 description 1
- 201000000459 head and neck squamous cell carcinoma Diseases 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 1
- 231100000844 hepatocellular carcinoma Toxicity 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
- 229920006158 high molecular weight polymer Polymers 0.000 description 1
- 210000001320 hippocampus Anatomy 0.000 description 1
- 210000003630 histaminocyte Anatomy 0.000 description 1
- 229960001330 hydroxycarbamide Drugs 0.000 description 1
- 230000036031 hyperthermia Effects 0.000 description 1
- 229940015872 ibandronate Drugs 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 238000003119 immunoblot Methods 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 230000006054 immunological memory Effects 0.000 description 1
- 238000012744 immunostaining Methods 0.000 description 1
- 238000009169 immunotherapy Methods 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- DBIGHPPNXATHOF-UHFFFAOYSA-N improsulfan Chemical compound CS(=O)(=O)OCCCNCCCOS(C)(=O)=O DBIGHPPNXATHOF-UHFFFAOYSA-N 0.000 description 1
- 229950008097 improsulfan Drugs 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 230000002458 infectious effect Effects 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 238000011221 initial treatment Methods 0.000 description 1
- 238000002743 insertional mutagenesis Methods 0.000 description 1
- QROONAIPJKQFMC-UHFFFAOYSA-N intoplicine Chemical compound C1=C(O)C=CC2=C(C=3C(NCCCN(C)C)=NC=C(C)C=3N3)C3=CC=C21 QROONAIPJKQFMC-UHFFFAOYSA-N 0.000 description 1
- 229950005428 intoplicine Drugs 0.000 description 1
- 230000010189 intracellular transport Effects 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- 229960004768 irinotecan Drugs 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 238000005304 joining Methods 0.000 description 1
- 229960003299 ketamine Drugs 0.000 description 1
- 229950005692 larotaxel Drugs 0.000 description 1
- 210000000867 larynx Anatomy 0.000 description 1
- 229940115286 lentinan Drugs 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 230000001665 lethal effect Effects 0.000 description 1
- GFIJNRVAKGFPGQ-LIJARHBVSA-N leuprolide Chemical compound CCNC(=O)[C@@H]1CCCN1C(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H]1NC(=O)CC1)CC1=CC=C(O)C=C1 GFIJNRVAKGFPGQ-LIJARHBVSA-N 0.000 description 1
- 229960004338 leuprorelin Drugs 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 239000012160 loading buffer Substances 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 230000005923 long-lasting effect Effects 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
- 239000000314 lubricant Substances 0.000 description 1
- 201000005249 lung adenocarcinoma Diseases 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 210000004324 lymphatic system Anatomy 0.000 description 1
- 210000005210 lymphoid organ Anatomy 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 229920002521 macromolecule Polymers 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 230000014759 maintenance of location Effects 0.000 description 1
- MQXVYODZCMMZEM-ZYUZMQFOSA-N mannomustine Chemical compound ClCCNC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CNCCCl MQXVYODZCMMZEM-ZYUZMQFOSA-N 0.000 description 1
- 229950008612 mannomustine Drugs 0.000 description 1
- 108010082117 matrigel Proteins 0.000 description 1
- 230000035800 maturation Effects 0.000 description 1
- WKPWGQKGSOKKOO-RSFHAFMBSA-N maytansine Chemical compound CO[C@@H]([C@@]1(O)C[C@](OC(=O)N1)([C@H]([C@@H]1O[C@@]1(C)[C@@H](OC(=O)[C@H](C)N(C)C(C)=O)CC(=O)N1C)C)[H])\C=C\C=C(C)\CC2=CC(OC)=C(Cl)C1=C2 WKPWGQKGSOKKOO-RSFHAFMBSA-N 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 229950009246 mepitiostane Drugs 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 230000001394 metastastic effect Effects 0.000 description 1
- 206010061289 metastatic neoplasm Diseases 0.000 description 1
- VJRAUFKOOPNFIQ-TVEKBUMESA-N methyl (1r,2r,4s)-4-[(2r,4s,5s,6s)-5-[(2s,4s,5s,6s)-5-[(2s,4s,5s,6s)-4,5-dihydroxy-6-methyloxan-2-yl]oxy-4-hydroxy-6-methyloxan-2-yl]oxy-4-(dimethylamino)-6-methyloxan-2-yl]oxy-2-ethyl-2,5,7,10-tetrahydroxy-6,11-dioxo-3,4-dihydro-1h-tetracene-1-carboxylat Chemical compound O([C@H]1[C@@H](O)C[C@@H](O[C@H]1C)O[C@H]1[C@H](C[C@@H](O[C@H]1C)O[C@H]1C[C@]([C@@H](C2=CC=3C(=O)C4=C(O)C=CC(O)=C4C(=O)C=3C(O)=C21)C(=O)OC)(O)CC)N(C)C)[C@H]1C[C@H](O)[C@H](O)[C@H](C)O1 VJRAUFKOOPNFIQ-TVEKBUMESA-N 0.000 description 1
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 102000021160 microtubule binding proteins Human genes 0.000 description 1
- 108091011150 microtubule binding proteins Proteins 0.000 description 1
- 230000029115 microtubule polymerization Effects 0.000 description 1
- XIVMHSNIQAICTR-UQYHODNASA-N milataxel Chemical compound O([C@H]1[C@@H]2[C@]3(OC(C)=O)CO[C@@H]3C[C@@H]([C@]2(C(=O)[C@H](O)C2=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=3OC=CC=3)C[C@]1(O)C2(C)C)C)OC(=O)CC)C(=O)C1=CC=CC=C1 XIVMHSNIQAICTR-UQYHODNASA-N 0.000 description 1
- 229960003539 mitoguazone Drugs 0.000 description 1
- MXWHMTNPTTVWDM-NXOFHUPFSA-N mitoguazone Chemical compound NC(N)=N\N=C(/C)\C=N\N=C(N)N MXWHMTNPTTVWDM-NXOFHUPFSA-N 0.000 description 1
- VFKZTMPDYBFSTM-GUCUJZIJSA-N mitolactol Chemical compound BrC[C@H](O)[C@@H](O)[C@@H](O)[C@H](O)CBr VFKZTMPDYBFSTM-GUCUJZIJSA-N 0.000 description 1
- 229950010913 mitolactol Drugs 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 229960000350 mitotane Drugs 0.000 description 1
- 239000003147 molecular marker Substances 0.000 description 1
- ZDZOTLJHXYCWBA-BSEPLHNVSA-N molport-006-823-826 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-BSEPLHNVSA-N 0.000 description 1
- 229910000403 monosodium phosphate Inorganic materials 0.000 description 1
- 235000019799 monosodium phosphate Nutrition 0.000 description 1
- FOYWNSCCNCUEPU-UHFFFAOYSA-N mopidamol Chemical compound C12=NC(N(CCO)CCO)=NC=C2N=C(N(CCO)CCO)N=C1N1CCCCC1 FOYWNSCCNCUEPU-UHFFFAOYSA-N 0.000 description 1
- 229950010718 mopidamol Drugs 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 229940124303 multikinase inhibitor Drugs 0.000 description 1
- 229960000951 mycophenolic acid Drugs 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- 208000017869 myelodysplastic/myeloproliferative disease Diseases 0.000 description 1
- 208000025113 myeloid leukemia Diseases 0.000 description 1
- NJSMWLQOCQIOPE-OCHFTUDZSA-N n-[(e)-[10-[(e)-(4,5-dihydro-1h-imidazol-2-ylhydrazinylidene)methyl]anthracen-9-yl]methylideneamino]-4,5-dihydro-1h-imidazol-2-amine Chemical compound N1CCN=C1N\N=C\C(C1=CC=CC=C11)=C(C=CC=C2)C2=C1\C=N\NC1=NCCN1 NJSMWLQOCQIOPE-OCHFTUDZSA-N 0.000 description 1
- 229940086322 navelbine Drugs 0.000 description 1
- 230000017074 necrotic cell death Effects 0.000 description 1
- 208000007538 neurilemmoma Diseases 0.000 description 1
- 208000015122 neurodegenerative disease Diseases 0.000 description 1
- 230000016273 neuron death Effects 0.000 description 1
- 229960002653 nilutamide Drugs 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
- 229960001420 nimustine Drugs 0.000 description 1
- VFEDRRNHLBGPNN-UHFFFAOYSA-N nimustine Chemical compound CC1=NC=C(CNC(=O)N(CCCl)N=O)C(N)=N1 VFEDRRNHLBGPNN-UHFFFAOYSA-N 0.000 description 1
- YMVWGSQGCWCDGW-UHFFFAOYSA-N nitracrine Chemical compound C1=CC([N+]([O-])=O)=C2C(NCCCN(C)C)=C(C=CC=C3)C3=NC2=C1 YMVWGSQGCWCDGW-UHFFFAOYSA-N 0.000 description 1
- 229950008607 nitracrine Drugs 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 210000004492 nuclear pore Anatomy 0.000 description 1
- 238000012758 nuclear staining Methods 0.000 description 1
- 238000001668 nucleic acid synthesis Methods 0.000 description 1
- 201000002575 ocular melanoma Diseases 0.000 description 1
- CZDBNBLGZNWKMC-MWQNXGTOSA-N olivomycin Chemical class O([C@@H]1C[C@@H](O[C@H](C)[C@@H]1O)OC=1C=C2C=C3C[C@H]([C@@H](C(=O)C3=C(O)C2=C(O)C=1)O[C@H]1O[C@@H](C)[C@H](O)[C@@H](OC2O[C@@H](C)[C@H](O)[C@@H](O)C2)C1)[C@H](OC)C(=O)[C@@H](O)[C@@H](C)O)[C@H]1C[C@H](O)[C@H](OC)[C@H](C)O1 CZDBNBLGZNWKMC-MWQNXGTOSA-N 0.000 description 1
- 229950011093 onapristone Drugs 0.000 description 1
- 229950001094 ortataxel Drugs 0.000 description 1
- 201000008968 osteosarcoma Diseases 0.000 description 1
- 230000036542 oxidative stress Effects 0.000 description 1
- VREZDOWOLGNDPW-UHFFFAOYSA-N pancratistatine Natural products C1=C2C3C(O)C(O)C(O)C(O)C3NC(=O)C2=C(O)C2=C1OCO2 VREZDOWOLGNDPW-UHFFFAOYSA-N 0.000 description 1
- 229940055729 papain Drugs 0.000 description 1
- 235000019834 papain Nutrition 0.000 description 1
- 229920002866 paraformaldehyde Polymers 0.000 description 1
- 244000052769 pathogen Species 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 229960002340 pentostatin Drugs 0.000 description 1
- QIMGFXOHTOXMQP-GFAGFCTOSA-N peplomycin 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)NCCCN[C@@H](C)C=1C=CC=CC=1)[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=1NC=NC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C QIMGFXOHTOXMQP-GFAGFCTOSA-N 0.000 description 1
- 229950003180 peplomycin Drugs 0.000 description 1
- 239000000816 peptidomimetic Substances 0.000 description 1
- 210000001428 peripheral nervous system Anatomy 0.000 description 1
- 210000001322 periplasm Anatomy 0.000 description 1
- 230000008823 permeabilization Effects 0.000 description 1
- 238000002823 phage display Methods 0.000 description 1
- 210000004214 philadelphia chromosome Anatomy 0.000 description 1
- 150000004713 phosphodiesters Chemical class 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- 239000002504 physiological saline solution Substances 0.000 description 1
- NUKCGLDCWQXYOQ-UHFFFAOYSA-N piposulfan Chemical compound CS(=O)(=O)OCCC(=O)N1CCN(C(=O)CCOS(C)(=O)=O)CC1 NUKCGLDCWQXYOQ-UHFFFAOYSA-N 0.000 description 1
- 229950001100 piposulfan Drugs 0.000 description 1
- 229960001221 pirarubicin Drugs 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 238000007747 plating Methods 0.000 description 1
- 150000003057 platinum Chemical class 0.000 description 1
- 230000004983 pleiotropic effect Effects 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 239000001103 potassium chloride Substances 0.000 description 1
- WCUXLLCKKVVCTQ-UHFFFAOYSA-M potassium chloride Inorganic materials [Cl-].[K+] WCUXLLCKKVVCTQ-UHFFFAOYSA-M 0.000 description 1
- 229960004694 prednimustine Drugs 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 230000001566 pro-viral effect 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
- 201000002212 progressive supranuclear palsy Diseases 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- VYXXMAGSIYIYGD-NWAYQTQBSA-N propan-2-yl 2-[[[(2R)-1-(6-aminopurin-9-yl)propan-2-yl]oxymethyl-(pyrimidine-4-carbonylamino)phosphoryl]amino]-2-methylpropanoate Chemical compound CC(C)OC(=O)C(C)(C)NP(=O)(CO[C@H](C)Cn1cnc2c(N)ncnc12)NC(=O)c1ccncn1 VYXXMAGSIYIYGD-NWAYQTQBSA-N 0.000 description 1
- 230000004952 protein activity Effects 0.000 description 1
- 108020001580 protein domains Proteins 0.000 description 1
- 230000017854 proteolysis Effects 0.000 description 1
- WOLQREOUPKZMEX-UHFFFAOYSA-N pteroyltriglutamic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)NC(CCC(=O)NC(CCC(=O)NC(CCC(O)=O)C(O)=O)C(O)=O)C(O)=O)C=C1 WOLQREOUPKZMEX-UHFFFAOYSA-N 0.000 description 1
- 101150010682 rad50 gene Proteins 0.000 description 1
- 150000003254 radicals Chemical class 0.000 description 1
- 229910052705 radium Inorganic materials 0.000 description 1
- HCWPIIXVSYCSAN-UHFFFAOYSA-N radium atom Chemical compound [Ra] HCWPIIXVSYCSAN-UHFFFAOYSA-N 0.000 description 1
- 229960002185 ranimustine Drugs 0.000 description 1
- BMKDZUISNHGIBY-UHFFFAOYSA-N razoxane Chemical compound C1C(=O)NC(=O)CN1C(C)CN1CC(=O)NC(=O)C1 BMKDZUISNHGIBY-UHFFFAOYSA-N 0.000 description 1
- 229960000460 razoxane Drugs 0.000 description 1
- 239000003642 reactive oxygen metabolite Substances 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 239000006215 rectal suppository Substances 0.000 description 1
- 229940100618 rectal suppository Drugs 0.000 description 1
- 210000000664 rectum Anatomy 0.000 description 1
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 description 1
- 230000013878 renal filtration Effects 0.000 description 1
- 238000009877 rendering Methods 0.000 description 1
- 238000003571 reporter gene assay Methods 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 238000002271 resection Methods 0.000 description 1
- 108091008146 restriction endonucleases Proteins 0.000 description 1
- 201000006845 reticulosarcoma Diseases 0.000 description 1
- 208000029922 reticulum cell sarcoma Diseases 0.000 description 1
- 229930002330 retinoic acid Natural products 0.000 description 1
- 230000001177 retroviral effect Effects 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 238000012552 review Methods 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
- 229950004892 rodorubicin Drugs 0.000 description 1
- MBABCNBNDNGODA-WPZDJQSSSA-N rolliniastatin 1 Natural products O1[C@@H]([C@@H](O)CCCCCCCCCC)CC[C@H]1[C@H]1O[C@@H]([C@H](O)CCCCCCCCCC[C@@H](O)CC=2C(O[C@@H](C)C=2)=O)CC1 MBABCNBNDNGODA-WPZDJQSSSA-N 0.000 description 1
- VHXNKPBCCMUMSW-FQEVSTJZSA-N rubitecan Chemical compound C1=CC([N+]([O-])=O)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 VHXNKPBCCMUMSW-FQEVSTJZSA-N 0.000 description 1
- 229930182947 sarcodictyin Natural products 0.000 description 1
- 238000012163 sequencing technique Methods 0.000 description 1
- 230000009919 sequestration Effects 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 239000002924 silencing RNA Substances 0.000 description 1
- 230000005783 single-strand break Effects 0.000 description 1
- 238000002741 site-directed mutagenesis Methods 0.000 description 1
- 229950001403 sizofiran Drugs 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 229960003787 sorafenib Drugs 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- ICXJVZHDZFXYQC-UHFFFAOYSA-N spongistatin 1 Natural products OC1C(O2)(O)CC(O)C(C)C2CCCC=CC(O2)CC(O)CC2(O2)CC(OC)CC2CC(=O)C(C)C(OC(C)=O)C(C)C(=C)CC(O2)CC(C)(O)CC2(O2)CC(OC(C)=O)CC2CC(=O)OC2C(O)C(CC(=C)CC(O)C=CC(Cl)=C)OC1C2C ICXJVZHDZFXYQC-UHFFFAOYSA-N 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 238000003153 stable transfection Methods 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 238000000528 statistical test Methods 0.000 description 1
- 238000002719 stereotactic radiosurgery Methods 0.000 description 1
- 230000001954 sterilising effect Effects 0.000 description 1
- 238000004659 sterilization and disinfection Methods 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 230000035882 stress Effects 0.000 description 1
- 229960001796 sunitinib Drugs 0.000 description 1
- WINHZLLDWRZWRT-ATVHPVEESA-N sunitinib Chemical compound CCN(CC)CCNC(=O)C1=C(C)NC(\C=C/2C3=CC(F)=CC=C3NC\2=O)=C1C WINHZLLDWRZWRT-ATVHPVEESA-N 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 206010042863 synovial sarcoma Diseases 0.000 description 1
- 230000009897 systematic effect Effects 0.000 description 1
- 229960001603 tamoxifen Drugs 0.000 description 1
- 229960004964 temozolomide Drugs 0.000 description 1
- NRUKOCRGYNPUPR-QBPJDGROSA-N teniposide 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](OC[C@H]4O3)C=3SC=CC=3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 NRUKOCRGYNPUPR-QBPJDGROSA-N 0.000 description 1
- 229960001278 teniposide Drugs 0.000 description 1
- 208000001608 teratocarcinoma Diseases 0.000 description 1
- 229950009016 tesetaxel Drugs 0.000 description 1
- 201000003120 testicular cancer Diseases 0.000 description 1
- 208000002918 testicular germ cell tumor Diseases 0.000 description 1
- 210000001550 testis Anatomy 0.000 description 1
- BPEWUONYVDABNZ-DZBHQSCQSA-N testolactone Chemical compound O=C1C=C[C@]2(C)[C@H]3CC[C@](C)(OC(=O)CC4)[C@@H]4[C@@H]3CCC2=C1 BPEWUONYVDABNZ-DZBHQSCQSA-N 0.000 description 1
- 229960005353 testolactone Drugs 0.000 description 1
- 230000004797 therapeutic response Effects 0.000 description 1
- 230000008719 thickening Effects 0.000 description 1
- 239000002562 thickening agent Substances 0.000 description 1
- 108060008226 thioredoxin Proteins 0.000 description 1
- 229940094937 thioredoxin Drugs 0.000 description 1
- 125000000341 threoninyl group Chemical group [H]OC([H])(C([H])([H])[H])C([H])(N([H])[H])C(*)=O 0.000 description 1
- YFTWHEBLORWGNI-UHFFFAOYSA-N tiamiprine Chemical compound CN1C=NC([N+]([O-])=O)=C1SC1=NC(N)=NC2=C1NC=N2 YFTWHEBLORWGNI-UHFFFAOYSA-N 0.000 description 1
- 229950011457 tiamiprine Drugs 0.000 description 1
- 239000003104 tissue culture media Substances 0.000 description 1
- 230000019432 tissue death Effects 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 229940044693 topoisomerase inhibitor Drugs 0.000 description 1
- XFCLJVABOIYOMF-QPLCGJKRSA-N toremifene Chemical compound C1=CC(OCCN(C)C)=CC=C1C(\C=1C=CC=CC=1)=C(\CCCl)C1=CC=CC=C1 XFCLJVABOIYOMF-QPLCGJKRSA-N 0.000 description 1
- 229960005026 toremifene Drugs 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- 238000003146 transient transfection Methods 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- 229960001727 tretinoin Drugs 0.000 description 1
- 150000004654 triazenes Chemical class 0.000 description 1
- 229960004560 triaziquone Drugs 0.000 description 1
- PXSOHRWMIRDKMP-UHFFFAOYSA-N triaziquone Chemical compound O=C1C(N2CC2)=C(N2CC2)C(=O)C=C1N1CC1 PXSOHRWMIRDKMP-UHFFFAOYSA-N 0.000 description 1
- 229930013292 trichothecene Natural products 0.000 description 1
- 150000003327 trichothecene derivatives Chemical class 0.000 description 1
- 230000001960 triggered effect Effects 0.000 description 1
- 229960001670 trilostane Drugs 0.000 description 1
- KVJXBPDAXMEYOA-CXANFOAXSA-N trilostane Chemical compound OC1=C(C#N)C[C@]2(C)[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CC[C@@]32O[C@@H]31 KVJXBPDAXMEYOA-CXANFOAXSA-N 0.000 description 1
- NOYPYLRCIDNJJB-UHFFFAOYSA-N trimetrexate Chemical compound COC1=C(OC)C(OC)=CC(NCC=2C(=C3C(N)=NC(N)=NC3=CC=2)C)=C1 NOYPYLRCIDNJJB-UHFFFAOYSA-N 0.000 description 1
- 229960001099 trimetrexate Drugs 0.000 description 1
- 229950000212 trioxifene Drugs 0.000 description 1
- 229960000875 trofosfamide Drugs 0.000 description 1
- UMKFEPPTGMDVMI-UHFFFAOYSA-N trofosfamide Chemical compound ClCCN(CCCl)P1(=O)OCCCN1CCCl UMKFEPPTGMDVMI-UHFFFAOYSA-N 0.000 description 1
- HDZZVAMISRMYHH-LITAXDCLSA-N tubercidin Chemical compound C1=CC=2C(N)=NC=NC=2N1[C@@H]1O[C@@H](CO)[C@H](O)[C@H]1O HDZZVAMISRMYHH-LITAXDCLSA-N 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 230000005740 tumor formation Effects 0.000 description 1
- 238000010396 two-hybrid screening Methods 0.000 description 1
- 229950009811 ubenimex Drugs 0.000 description 1
- 238000010798 ubiquitination Methods 0.000 description 1
- 238000005199 ultracentrifugation Methods 0.000 description 1
- 238000009281 ultraviolet germicidal irradiation Methods 0.000 description 1
- 208000018417 undifferentiated high grade pleomorphic sarcoma of bone Diseases 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- AHXICHPPXIGCBN-GPWPDEGDSA-N uqc681jjiv Chemical compound O([C@H]1[C@H]2[C@@](C([C@H](OC(C)=O)C3=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C(C)(C)C)C[C@]1(O)C3(C)C)=O)(C)[C@@H](O)C[C@H]1OC[C@]12OC(=O)OC)C(=O)C1=CC=CC=C1 AHXICHPPXIGCBN-GPWPDEGDSA-N 0.000 description 1
- DNYWZCXLKNTFFI-UHFFFAOYSA-N uranium Chemical compound [U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U][U] DNYWZCXLKNTFFI-UHFFFAOYSA-N 0.000 description 1
- 206010046766 uterine cancer Diseases 0.000 description 1
- 238000001291 vacuum drying Methods 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
- 229960004355 vindesine Drugs 0.000 description 1
- UGGWPQSBPIFKDZ-KOTLKJBCSA-N vindesine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(N)=O)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1N=C1[C]2C=CC=C1 UGGWPQSBPIFKDZ-KOTLKJBCSA-N 0.000 description 1
- CILBMBUYJCWATM-PYGJLNRPSA-N vinorelbine ditartrate Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O.OC(=O)[C@H](O)[C@@H](O)C(O)=O.C1N(CC=2C3=CC=CC=C3NC=22)CC(CC)=C[C@H]1C[C@]2(C(=O)OC)C1=CC([C@]23[C@H]([C@@]([C@H](OC(C)=O)[C@]4(CC)C=CCN([C@H]34)CC2)(O)C(=O)OC)N2C)=C2C=C1OC CILBMBUYJCWATM-PYGJLNRPSA-N 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 238000012800 visualization Methods 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 230000003442 weekly effect Effects 0.000 description 1
- 229940053867 xeloda Drugs 0.000 description 1
- BPICBUSOMSTKRF-UHFFFAOYSA-N xylazine Chemical compound CC1=CC=CC(C)=C1NC1=NCCCS1 BPICBUSOMSTKRF-UHFFFAOYSA-N 0.000 description 1
- 229960001600 xylazine Drugs 0.000 description 1
- 229950009268 zinostatin Drugs 0.000 description 1
- 229960000641 zorubicin Drugs 0.000 description 1
- FBTUMDXHSRTGRV-ALTNURHMSA-N zorubicin 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)=N\NC(=O)C=1C=CC=CC=1)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 FBTUMDXHSRTGRV-ALTNURHMSA-N 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/1703—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- A61K38/1709—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- A61K38/1716—Amyloid plaque core protein
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/14—Type of nucleic acid interfering N.A.
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2320/00—Applications; Uses
- C12N2320/30—Special therapeutic applications
- C12N2320/31—Combination therapy
Definitions
- the present invention is in the field of medicine, in particular oncology, for decreasing therapeutic acquired resistance to chemotherapy and/or radiotherapy in a subject.
- Double-strand DNA breaks are considered as the most lethal DNA lesions and are commonly induced in cancer radio- or chemo-therapy.
- DSB-associated therapies is the tumor resistance developed before or after treatment [1,2]. Thus, it will be important to decipher the underlying molecular mechanism of this resistance to improve existing regimens.
- DSBs activate DNA damage response pathways through the kinases ATM, ATR and DNA-PKcs, that in turn activate proteins involved in two main pathways, the classical nonhomologous end-joining (cNHEJ) and the homologous recombination (HR).
- cNHEJ mediates repair by directly rejoining DSB ends, contrary to HR which utilizes a homologous DNA sequence to guide DNA repair [3,4], Which of the two repair routes is used depends in part of the cell cycle phase. Since HR relies on the presence of a sister chromatid, it can be effective only in the late S/G2 phase while the NHEJ pathway is effective throughout the cell cycle.
- microtubule dynamics was demonstrated to be crucial in the intracellular trafficking of DNA repair proteins [9]
- Tau was first described as a neuronal microtubule-associated protein (MAPT) and when it aggregates it is a major player in neurodegenerative diseases like Alzheimer’s [11]
- AGT neuronal microtubule-associated protein
- Tau has many other cellular functions [12]
- it interferes with several biological processes such as signaling pathways, RNA metabolism and it even contributes to the inflammatory response.
- Tau is not only found in the cytoplasm but also in the cell nucleus where it participates in chromatin organisation in neuronal and cancer cells [13,14].
- Tau deletion induces an increase in DSBs and a slower DNA repair in neurons after reactive oxygen species induction triggered by hyperthermia [16,17],
- the molecular mechanism remains elusive.
- Tau expression in cancer cells could be involved in resistance to conventional anti-cancer treatments, in particular those inducing DNA damage.
- the present invention is in the field of medicine, in particular oncology, for decreasing therapeutic acquired resistance to chemotherapy and/or radiotherapy in a subject.
- the present invention is defined by the claims.
- Therapeutic resistance is one of the major challenges in cancer treatment. These latter may be inherently resistant or this resistance may be acquired during treatment.
- the tau protein encoded by the MAPT gene, could prove to be an important contributor in resistance to cancer therapy.
- a literature abundant has now demonstrated that the expression of his gene is increased in several types of cancers and could be associated with resistance to taxanes. This effect has been associated with its well- known role regulator of microtubule assembly and disassembly.
- tau was a pleiotropic protein, involved in several mechanisms cellular. Among them, its role in genome protection/repair could be directly related to resistance to other anti-cancer treatments, especially those inducing DNA damage.
- the inventors show that DSBs from DNA disappear faster after irradiation or bleomycin treatment in MCF7 and MDA-MB-231 cells expressing MAPT in a manner endogenous than in their homologs stably transfected with a tau-directed shRNA (shTau).
- the inventors have been thus abled to confirm in vivo that the tau protein was a factor of resistance to radiotherapy and the chemotherapy either inducing DSB (doxorubicin) or DNA adducts (cisplatin and oxaliplatin) and that the reduction of tau expression by shRNA increased the sensitivity of tumors to these treatments.
- DSB doxorubicin
- DNA adducts cisplatin and oxaliplatin
- the present invention relates to a method for decreasing therapeutic acquired resistance to chemotherapy agent and/or radiotherapy agent in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a Tau inhibitor.
- the present invention relates to a method for preventing and/or treating cancer with acquired resistance to treatment with chemotherapy agent and/or radiotherapy agent in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a Tau inhibitor
- a patient denotes a mammal, such as a rodent, a feline, a canine, and a primate.
- a patient according to the invention is a human.
- a patient according to the invention refers to any subject (preferably human) afflicted with or susceptible to be afflicted with a cancer.
- DNA repair refers to a collection of processes by which a cell identifies and corrects damage to DNA molecules. Single-strand defects are repaired by base excision repair (BER), nucleotide excision repair (NER), or mismatch repair (MMR). Double-strand breaks are repaired by non-homologous end joining (NHEJ), microhomology- mediated end joining (MMEJ), or homologous recombination.
- NHEJ non-homologous end joining
- MMEJ microhomology- mediated end joining
- homologous recombination After DNA damage, cell cycle checkpoints are activated, which pause the cell cycle to give the cell time to repair the damage before continuing to divide.
- Checkpoint mediator proteins include BRCA1, MDC1, 53BP1, p53, ATM, ATR, CHK1, CHK2, and p21.
- the term "impaired DNA repair” refers to a state in which a mutated cell or a cell with altered gene expression is incapable of DNA repair or has reduced activity of one or more DNA repair pathways or takes longer to repair damage to its DNA as compared to a wild type cell.
- cancer refers to or describe the pathological condition in mammals that is typically characterized by unregulated cell growth. More precisely, in the methods of the invention, diseases, namely tumors that do express/secrete Tau protein are most likely to respond to the Tau inhibitor treatment. In particular, the cancer may be associated with a solid tumor or lymphoma/leukemia (from hematopoietic cell).
- cancers that are associated with solid tumor formation include breast cancer, uterine/cervical cancer, oesophageal cancer, pancreatic cancer, colon cancer, colorectal cancer, kidney cancer, ovarian cancer, prostate cancer, head and neck cancer, non small cell lung cancer stomach cancer, tumors of mesenchymal origin (i.e; fibrosarcoma and rhabdomyoscarcoma) thyroid cancer.
- cancers include chronic myeloid leukemia, acute lymphoblastic leukemia, Philadelphia chromosome positive acute lymphoblastic leukemia (Ph+ ALL), squamous cell carcinoma, small-cell lung cancer, non-small cell lung cancer, glioma, gastrointestinal cancer, renal cancer, ovarian cancer, liver cancer, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, stomach cancer, bladder cancer, hepatocarcinoma, breast cancer, colon carcinoma, and head and neck cancer, gastric cancer, germ cell tumor, pediatric sarcoma, multiple myeloma, acute myelogenous leukemia (AML), chronic lymphocytic leukemia, mastocytosis and any symptom associated with mastocytosis.
- AML acute myelogenous leukemia
- carcinoma including that of the bladder (including accelerated and metastatic bladder cancer), breast, colon (including colorectal cancer), kidney, liver, lung (including small and non-small cell lung cancer and lung adenocarcinoma), ovary, prostate, testis, genitourinary tract, lymphatic system, rectum, larynx, pancreas (including exocrine pancreatic carcinoma), esophagus, stomach, gall bladder, cervix, thyroid, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, Tcell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma, histiocytic lymphoma, and Burketts lymph
- the cancer is a hematopoietic cancer, especially a leukemia or lymphoma.
- the cancer is a solid tumor.
- the cancer may be sarcoma and oestosarcoma such as Kaposi sarcome, AIDS-related Kaposi sarcoma, melanoma, in particular ulveal melanoma, and cancers of the head and neck, kidney, ovary, pancreas, prostate, thyroid, lung, esophagus, breast, bladder, colorectum, liver and biliary tract, uterine, appendix, and cervix, testicular cancer, gastrointestinal cancers and endometrial and peritoneal cancers.
- the cancer may be sarcoma, melanoma, in particular ulveal melanoma, and cancers of the head and neck, kidney, ovary, pancreas, prostate, thyroid, lung, esophagus, breast, bladder, colorectum, liver, cervix, and endometrial and peritoneal cancers.
- the cancer may be selected from the group consisting of breast cancer, hepatocellular carcinoma, colorectal cancer, glioblastoma, melanoma, and head and neck cancer.
- the solid tumor is selected from the group consisting of breast cancer ((Rouzier et al., 2005;Matrone et al., 2010;Spicakova et al., 2010;Li et al., 2013), gastric cancer (Wang Q et al Pathol. Oncol. Res. (2013) 19:429-435), ovarian cancer (Smoter M. et al.
- NETs Neuroendocrine tumors
- DLB Double-strand breaks
- NHEJ nonhomologous DNA end joining
- NHEJ repair enzymes act iteratively, act in any order, and can function independently of one another at each of the two DNA ends being joined.
- NHEJ is critical not only for the repair of pathologic DSBs as in chromosomal translocations, but also for the repair of physiologic DSBs created during V(D)J recombination and class switch recombination. Therefore, patients lacking normal NHEJ are not only sensitive to ionizing radiation, but also severely immunodeficient
- Radiotherapy includes, but is not limited to, y-rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells.
- Other radiotherapies include microwaves and UV-irradiation.
- Other approaches to radiation therapy are also contemplated in the present invention.
- the DNA-damaging antitumor agent is preferably selected from the group consisting of an inhibitor of topoisomerases I or II, a DNA crosslinker, a DNA alkylating agent, an anti- metabolic agent and inhibitors of the mitotic spindles.
- Inhibitors of topoisomerases I and/or II include, but are not limited to, etoposide, topotecan, camptothecin, irinotecan, amsacrine, intoplicine, anthracyclines such as doxorubicin, epirubicine, daunorubicine, idanrubicine and mitoxantrone.
- Inhibitors of Topoisomerase I and II include, but are not limited to, intoplecin.
- the DNA-damaging antitumor agent is doxorubicin.
- DNA crosslinkers include, but are not limited to, cisplatin, carboplatin and oxaliplatin.
- the DNA-damaging antitumor agent is selected from the group consisting of carboplatin and oxaliplatin.
- Anti-metabolic agents block the enzymes responsible for nucleic acid synthesis or become incorporated into DNA, which produces an incorrect genetic code and leads to apoptosis.
- Nonexhaustive examples thereof include, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors, and more particularly Methotrexate, Floxuridine, Cytarabine, 6-Mercaptopurine, 6- Thioguanine, Fludarabine phosphate, Pentostatine, 5 -fluorouracil (5-FU), gemcitabine and capecitabine.
- the DNA-damaging anti-tumor agent can be alkylating agents including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas, metal salts and triazenes.
- Nonexhaustive examples thereof include Uracil mustard, Chlormethine, Cyclophosphamide (CYTOXAN(R)), Ifosfamide, Melphalan, Chlorambucil, Pipobroman, Triethylenemelamine, Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Fotemustine, cisplatin, carboplatin, oxaliplatin, thiotepa, Streptozocin, dacarbazine, and Temozolomide.
- Inhibitors of the mitotic spindles include, but are not limited to, , vinorelbine, larotaxel (also called XRP9881; Sanofi-Aventis), XRP6258 (Sanofi-Aventis), BMS-184476 (Bristol- Meyer-Squibb), BMS-188797 (Bristol-Meyer-Squibb), BMS-275183 (Bristol-Meyer-Squibb), ortataxel (also called IDN 5109, BAY 59-8862 or SB-T-101131 ; Bristol-Meyer-Squibb), RPR 109881 A (Bristol-Meyer-20 Squibb), RPR 116258 (Bristol-Meyer-Squibb), NBT-287 (TAPESTRY), Tesetaxel (also called DJ-927), IDN 5390 (INDENA), and MAC-321 (WYETH). Also see the review of Hennenfent & Govindan (2006, Anna
- the DNA-damaging antitumor agent is an inhibitor of topoisomerases I and/or II, a DNA crosslinker, an anti-metabolic agent or a combination thereof.
- the DNA damaging antitumor agent is selected from the group consisting of doxorubicin, 5-FU, carboplatin and oxaliplatin or a combination thereof.
- the conjugated DBait is DT01 and the DNA-damaging antitumor agent is selected from the group consisting of doxorubicin, carboplatin, 5-FU and oxaliplatin.
- the term “taxane” has its general meaning in the art and relates to a class of diterpenes. They were originally identified from plants of the genus Taxus (yews), and feature a taxadiene core. Paclitaxel (Taxol) and docetaxel (Taxotere) are widely used as chemotherapy agents. Cabazitaxel was FDA approved to treat hormone-refractory prostate cancer.
- the DNA-damaging antitumor agent is not a taxane agent. In some embodiment, the DNA-damaging antitumor agent is not the following taxane agent: oxetaxel, paclitaxel, abraxane.
- the taxane is not oxetaxel. In some embodiment, the taxane is not paclitaxel. In some embodiment, the taxane is not abraxane.
- the method of the present invention does not comprise the administration of DNA-damaging antitumor agent does not comprise a taxane agent. In some embodiment, the method of the present invention does not comprise the administration of oxetaxel, paclitaxel, abraxane.
- the DNA-damaging antitumor agent of the present invention does not comprise the taxane-like compounds described in Papin S, Paganetti P. Emerging Evidences for an Implication of the Neurodegeneration -Associated Protein TAU in Cancer. Brain Sci. 2020 Nov 16;10(l 1):862. doi: 10.3390/brainscilOl 10862.
- Tau in cancer cells could be involved in acquired resistance to conventional anti-cancer treatment, in particular those inducing DNA damage. In some embodiment, Tau in cancer cells could be involved in acquired resistance to radiotherapy or to chemotherapy.
- the term “acquired resistance” indicates that the cancer becomes resistant and/or substantially less response to the effects of the drug after being exposed to it for a certain period of time.
- the term “radiation therapy” or “radiotherapy” has its general meaning in the art and refers the treatment of cancer with ionizing radiation. Ionizing radiation deposits energy that injures or destroys cells in the area being treated (the target tissue) by damaging their genetic material, making it impossible for these cells to continue to grow.
- One type of radiation therapy commonly used involves photons, e.g. X-rays. Depending on the amount of energy they possess, the rays can be used to destroy cancer cells on the surface of or deeper in the body. The higher the energy of the x-ray beam, the deeper the x-rays can go into the target tissue. Linear accelerators and betatrons produce x-rays of increasingly greater energy.
- Gamma rays are another form of photons used in radiation therapy. Gamma rays are produced spontaneously as certain elements (such as radium, uranium, and cobalt 60) release radiation as they decompose, or decay.
- the radiation therapy is external radiation therapy.
- external radiation therapy examples include, but are not limited to, conventional external beam radiation therapy; three-dimensional conformal radiation therapy (3D-CRT), which delivers shaped beams to closely fit the shape of a tumor from different directions; intensity modulated radiation therapy (IMRT), e.g., helical tomotherapy, which shapes the radiation beams to closely fit the shape of a tumor and also alters the radiation dose according to the shape of the tumor; conformal proton beam radiation therapy; image-guided radiation therapy (IGRT), which combines scanning and radiation technologies to provide real time images of a tumor to guide the radiation treatment; intraoperative radiation therapy (IORT), which delivers radiation directly to a tumor during surgery; stereotactic radiosurgery, which delivers a large, precise radiation dose to a small tumor area in a single session; hyperfractionated radiation therapy, e.g., continuous hyperfractionated accelerated radiation therapy (CHART), in which more than one treatment (fraction) of radiation therapy are given to a subject per day; and hypofractionated radiation therapy, in which larger doses of radiation therapy per fraction
- the present methods of invention invention relates to radiotherapy inducing double-strans breaks.
- ionizing radiations cause directly or indirectly double-stranded breaks (DSBs) and trigger cell/tissue death (necrosis or apoptosis).
- DSBs double-stranded breaks
- cytotoxic effect of ionizing radiation forms the basis for radiotherapy, which is widely used in the treatment of human cancer.
- the efficacy of radiotherapy is currently limited by the radio-resistance of certain tumors (for example, glioblastoma, head and neck squamous cell carcinoma) and by the side effects caused by irradiation of nearby normal tissues (for example, in the treatment of breast and cervical cancer).
- radiosensitivity refers to the relative susceptibility of cells to the harmful effect of ionizing radiation. The more radiosensitive a cell is, the less radiation that is required to kill that cell. In general, it has been found that cell radiosensitivity is directly proportional to the rate of cell division and inversely proportional to the cell's capacity for DNA repair.
- radioresistant refers to a cell that does not die when exposed to clinically suitable dosages of radiation.
- radiotoxicity designates any toxic or adverse side effect induced by radiation which may be observed in a subject treated by radiation therapy or in an organotypic slice which was irradiated.
- radiotoxicity includes any toxic or adverse side effect of radiation, which may be observed in the irradiated organotypic slice comprising healthy tissue.
- the radiotoxicity also includes any toxic or adverse side effect of radiation, which may be observed in the irradiated organotypic slice comprising healthy tissue and cancer tissue.
- chemotherapeutic agents include multikinase inhibitors such as sorafenib and sunitinib, alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, tri ethyl enethiophosphaorarni de and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; cally
- multikinase inhibitors such as sorafenib and sunitinib
- alkylating agents such as thi
- calicheamicin especially calicheamicin (11 and calicheamicin 211, see, e.g., Agnew Chem Inti. Ed. Engl. 33: 183-186 (1994); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, canninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholinodoxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino
- antihormonal agents that act to regulate or inhibit honnone action on tumors
- anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
- the present invention relates to chemotherapy inducing doublestranded breaks (DSBs).
- DSBs doublestranded breaks
- chemotherapeutic agents can cause DNA damages, including direct or indirect double-stranded breaks.
- chemotherapeutic agents are: inhibitors of topoisomerases I or II (camptothecin/topotecan, epirubicin/etoposide), DNA crosslinkers (cisplatin/carboplatin/oxaliplatin), DNA alkylating agents (carmustine/dacarbazine) or anti-metabolic agents (5- fluorouracil/gemcitabine/capecitabine).
- chemosensitivity refers to the relative susceptibility of cancer cells to the effects of anticancer drugs. The more chemosensitive a cancer cell is, the less anticancer drug is required to kill that cell.
- treatment refers to both prophylactic or preventive treatment as well as curative, improving the patient’s condition or disease modifying treatment, including treatment of patient at risk of contracting the disease or suspected to have contracted the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse.
- the treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment.
- therapeutic regimen is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy.
- a therapeutic regimen may include an induction regimen and a maintenance regimen.
- the phrase “induction regimen” or “induction period” refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease.
- the general goal of an induction regimen is to provide a high level of drug to a patient during the initial period of a treatment regimen.
- An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both.
- maintenance regimen refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a patient during treatment of an illness, e.g., to keep the patient in remission for long periods of time (months or years).
- a maintenance regimen may employ continuous therapy (e.g., administering a drug at regular intervals, e.g., daily, weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., disease manifestation, etc.]).
- the term “preventing” intends characterizing a prophylactic method or process that is aimed at delaying or preventing the onset of a disorder or condition to which such term applies.
- a gene product can be the direct transcriptional product of a gene (e.g., mRNA, tRNA, rRNA, antisense RNA, ribozyme, structural RNA or any other type of RNA) or a protein (i.e. Tau) produced by translation of a mRNA.
- a gene product can be the direct transcriptional product of a gene (e.g., mRNA, tRNA, rRNA, antisense RNA, ribozyme, structural RNA or any other type of RNA) or a protein (i.e. Tau) produced by translation of a mRNA.
- Tau denotes the Tau protein from mammals and especially from primates (and Tupaiidae).
- Human Tau is a neuronal microtubule-associated protein found predominantly in axons and functions to promote tubulin polymerization and stabilize microtubules.
- Six isoforms are found in the human brain, the longest isoform comprising 441 amino acids (isoform F, Uniprot P10636-8).
- Tau and its properties are also described by Reynolds, C. H. et al., J. Neurochem. 69 (1997) 191-198.
- Tau in its hyperphosphorylated form, is the major component of paired helical filaments (PHF), the building block of neurofibrillary lesions in Alzheimer's disease (AD) brain.
- PHF paired helical filaments
- AD Alzheimer's disease
- Tau can be phosphorylated at its serine or threonine residues by several different kinases including GSK3beta, cdk5, MARK and members of the MAP kinase family.
- the protein sequence of human Tau protein, and its isoforms, may be found in Uniprot database with the following access numbers:
- human Tau protein is encoded by the MAPT (Microtubule associated protein tau) gene located on chromosome 17 (Gene ID: 4137). This gene has 18 transcripts (splice variants), 1 gene allele, 255 orthologues, 1 paralogue and is associated with 13 phenotypes.
- MAPT Microtubule associated protein tau
- Example of human MAPT transcripts which encoded Tau protein may be found in Ensembl database with the following access number
- Transcript MAPT-201 (833 AA) Ensembl ID ENST00000262410 (Protein coding)
- Transcript MAPT-202 (352 AA) Ensembl ID ENST00000334239 (Protein coding Transcript MAPT-203(736 AA) Ensembl ID ENST00000344290 (Protein coding) Transcript MAPT-204 (441 AA) Ensembl ID ENST00000351559 (Protein coding) Transcript MAPT-205 (776 AA) Ensembl ID ENST00000415613 (Protein coding) Transcript MAPT-206 (412 AA) Ensembl ID :ENST00000420682 (Protein coding) Transcript MAPT-207 (410 AA) Ensembl ID ENST00000431008 (Protein coding) Transcript MAPT-208 (383 AA) Ensembl ID ENST00000446361 (Protein coding)
- Transcipt MAPT-209 (381 AA) Ensembl ID ENST00000535772 (Protein coding) Transcript MAPT-212 (758 AA) Ensembl ID ENST00000571987 (Protein coding) Transript MAPT-214 (441 AA) Ensembl ID ENST00000574436 (Protein coding) Transcript MAPT-217 (412 AA) Ensembl ID ENST00000680542 (Protein coding) Transcript MAPT-218 (424 AA) Ensembl ID ENST00000680674 (Protein coding)
- variant sequences of the Tau may be used in the context of the present invention, those including but not limited to functional homologues, paralogues or orthologues of such sequences.
- Tau should be understood broadly, it encompasses the native Tau, variants thereof having binding activity with microtubule and fragments thereof having binding activity with microtubule.
- native Tau, variants and isoforms preferably contain at least three or four microtubule binding domains (named 3R and 4R respectively). All human Tau isoform and MAPT transcript above described contains at least three or four microtubule binding domains.
- the Tau protein used in the context of the present invention is transcript Variant (or Tau Isoform) selected from the list consisting of (1N4R) Transcript MAPT-206 (412 AA) (Protein coding Tau isoform E) and (2N4R) Transript MAPT-214 (441 AA) (Protein coding Tau isoform F).
- transcript Variant or Tau Isoform
- the inhibitor of the present invention is Tau inhibitor.
- inhibitor includes not only drugs for inhibiting activity of target molecules, but also drugs for inhibiting the expression of target molecules.
- Tau inhibitor denotes a molecule or compound which can inhibit directly or indirectly the activity of the protein by limiting or impairing the interactions of the protein (ie with microtubules associated with translocation of DNA repair factors such as 53BP1 or with chromatin proteins and DNA), or a molecule or compound which destabilizes the protein structure, or a molecule or compound which inhibits the transcription or the translation of Tau, or accelerates its degradation.
- Tau inhibitor also denotes an inhibitor of the expression of the gene coding for the protein.
- the Tau inhibitor is a Tau inhibitor which directly binds to tau (protein or nucleic sequence (DNA or mRNA)) and neutralizes, blocks, inhibits, abrogates, reduces or interferes with the biological activity of Tau protein such as 1) Tau protein binding activity with microtubules (associated with translocation of DNA repair factors such as 53BP1) and/or 2) initiation of repair activities of the junction-type DSBs of the non-homologous ends (NHEJ) and homologous recombination (HR) .
- tau protein or nucleic sequence (DNA or mRNA)
- the Tau inhibitor (i) directly binds to Tau (protein or nucleic sequence (DNA or mRNA)) and (ii) inhibits biological activity of Tau protein such as Tau protein binding activity with microtubules (associated with translocation in nucleus of DNA repair factors such as 53BP1) and/or initiation of repair activities of the junction-type DSBs of the non-homologous ends (NHEJ) and homologous recombination (HR).
- Tau inhibitors include but are not limited to any of the inhibitors described in “Jadhav et al. Acta Neuropathologica Communications (2019) 7:22 all of which are herein incorporated by reference.
- biological activity of Tau protein is meant in the context of the present invention, the translocation in nucleus of DNA repair factors such as 53BP1) and/or initiation of repair activities of the junction-type DSBs of the non-homologous ends (NHEJ) and homologous recombination (HR)
- the antagonist/inhibitor specifically binds to Tau protein (protein or nucleic sequence (DNA or mRNA)) in a sufficient manner to inhibit the biological activity of Tau protein. Binding to Tau protein and inhibition of the biological activity of Tau protein may be determined by any competing assays well known in the art.
- the assay may consist in determining the ability of the agent to be tested as a Tau protein inhibitor to bind to Tau protein. The binding ability is reflected by the Kd measurement.
- Kd is intended to refer to the dissociation constant, which is obtained from the ratio of Kd to Ka (i.e.
- Kd/Ka Kd/Ka and is expressed as a molar concentration (M).
- Kd values for binding biomolecules can be determined using methods well established in the art.
- an antagonist / inhibitor that "specifically binds to Tau protein" is intended to refer to an inhibitor that binds to human Tau protein polypeptide with a Kd of IpM or less, lOOnM or less, lOnM or less, or 3nM or less.
- a competitive assay may be settled to determine the ability of the agent to inhibit biological activity of Tau protein : inhibition of Tau protein binding with microtubules (associated with translocation in nucleus of DNA repair factors such as 53BP1) and/or initiation of repair activities of the junction-type DSBs of the non-homologous ends (NHEJ) and homologous recombination (HR).
- microtubules associated with translocation in nucleus of DNA repair factors such as 53BP1
- inhibitor of the Tau protein activity it is herein referred to a compound which is capable of reducing or suppressing 1) the translocation in nucleus of DNA repair factors such as 53BP1 and/or 2) initiation of repair activities of the junction-type DSBs of the non- homologous ends (NHEJ) and homologous recombination (HR).
- a compound is an inhibitor 1) the translocation in nucleus of DNA repair factors such as 53BP1 and/or 2) initiation of repair activities of the junction-type DSBs of the non-homologous ends (NHEJ) and homologous recombination (HR).
- NHEJ non-homologous ends
- HR homologous recombination
- DSBs is quantified using a phosphorylated form of H2AX as an indicator of DNA damagefsee 29],
- a tau inhibitor according to the invention includes but is not limited to:
- A) Inhibitor of Tau activity selected from the list consisting Anti-Tau antibody and anti- Tau aptamers, tau peptide (vaccines)
- PROTAC Protein Transfer Targeting Chimera
- Inhibitor of Tau gene expression selected from the list consisting of antisense, oligonucleotide, nuclease, siRNA, shRNA or ribozyme nucleic acid sequence.
- the Tau inhibitor is an antibody.
- antibody is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, multi-specific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
- the term includes antibody fragments that comprise an antigen binding domain such as Fab', Fab, F(ab')2, single domain antibodies (DABs), TandAbs dimer, Fv, scFv (single chain Fv), dsFv, ds-scFv, Fd, linear antibodies, VHH (antigen binding fragment of heavy chain only antibodies), minibodies, diabodies, bi specific antibody fragments, bibody, tribody (scFv-Fab fusions, bispecific or trispecific, respectively); sc-diabody; kappa(lamda) bodies (scFv-CL fusions); BiTE (Bispecific T-cell Engager, scFv-scFv tandems to attract T cells); DVD-Ig (dual variable domain antibody, bispecific format); SIP (small immunoprotein, a kind of minibody); SMIP ("small modular immunopharmaceutical” scFv-Fc dimer; DART (ds-stabilized dia
- Antibodies can be fragmented using conventional techniques. For example, F(ab')2 fragments can be generated by treating the antibody with pepsin. The resulting F(ab')2 fragment can be treated to reduce disulfide bridges to produce Fab' fragments. Papain digestion can lead to the formation of Fab fragments.
- Fab, Fab' and F(ab')2, scFv, Fv, dsFv, Fd, dAbs, TandAbs, ds-scFv, dimers, minibodies, diabodies, bispecific antibody fragments and other fragments can also be synthesized by recombinant techniques or can be chemically synthesized. Techniques for producing antibody fragments are well known and described in the art. For example, each of Beckman et al., 2006; Holliger & Hudson, 2005; Le Gall et al., 2004; Reff & Heard, 2001 ; Reiter et al., 1996; and Young et al., 1995 further describe and enable the production of effective antibody fragments.
- the antibody is a “chimeric” antibody as described in U.S. Pat. No. 4,816,567.
- the antibody is a humanized antibody, such as described U.S. Pat. Nos. 6,982,321 and 7,087,409.
- the antibody is a human antibody.
- a “human antibody” such as described in US 6,075,181 and 6,150,584.
- the antibody is a single domain antibody such as described in EP 0 368 684, WO 06/030220 and WO 06/003388.
- the inhibitor is a monoclonal antibody.
- Monoclonal antibodies can be prepared and isolated using any technique to produce antibody molecules by continuous cell lines in culture. Techniques for production and isolation include but are not limited to the hybridoma technique, the human B-cell hybridoma technique and the EBV-hybridoma technique.
- the antibody is specific of the isoform B of Tau.
- Antibodies directed against Tau can be raised according to known methods by administering the appropriate antigen or epitope to a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others.
- a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others.
- Various adjuvants known in the art can be used to enhance antibody production.
- antibodies useful in practicing the invention can be polyclonal, monoclonal antibodies are preferred.
- Monoclonal antibodies against TAU can be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture.
- Techniques for production and isolation include but are not limited to the hybridoma technique originally described by Kohler and Milstein (1975); the human B-cell hybridoma technique (Cote et al., 1983); and the EBV-hybridoma technique (Cole et al. 1985).
- techniques described for the production of single chain antibodies can be adapted to produce anti-TAU single chain antibodies.
- Anti-TAU antibody fragments including but not limited to F(ab')2 fragments, which can be generated by pepsin digestion of an intact antibody molecule, and Fab fragments, which can be generated by reducing the disulfide bridges of the F(ab')2 fragments.
- Fab and/or scFv expression libraries can be constructed to allow rapid identification of fragments having the desired specificity to Tau.
- Humanized anti-TAU antibodies and antibody fragments therefrom can also be prepared according to known techniques. "Humanized antibodies” are forms of nonhuman (e.g., rodent) chimeric antibodies that contain minimal sequence derived from non- human immunoglobulin.
- humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (CDRs) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity.
- donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity.
- framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
- humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
- the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence.
- the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
- Fc immunoglobulin constant region
- Tau inhibitors such as anti Tau antibodies are well known in the art. Examples of patents disclosing anti Tau antibodies are. WO/2012/049570, WO/2014096321, WO/2015/004163; WO/2015200806, WO/2017/112078, WO/2018/152359, WO/2020/ 120644 (VHH anti Tau) WO/2020193520, WO/2021/010712,. . .
- the antibody according to the invention is a single domain antibody directed against Tau.
- the term “single domain antibody” (sdAb) or “VHH” refers to the single heavy chain variable domain of antibodies of the type that can be found in Camelid mammals which are naturally devoid of light chains. Such VHH are also called “nanobody®”. According to the invention, sdAb can particularly be llama sdAb.
- VHH refers to the single heavy chain having 3 complementarity determining regions (CDRs): CDR1, CDR2 and CDR3.
- CDR complementarity determining region
- VHH complementarity determining region
- the VHH according to the invention can readily be prepared by an ordinarily skilled artisan using routine experimentation.
- the VHH variants and modified form thereof may be produced under any known technique in the art such as in-vitro maturation.
- VHHs or sdAbs are usually generated by PCR cloning of the V-domain repertoire from blood, lymph node, or spleen cDNA obtained from immunized animals into a phage display vector, such as pHEN2.
- Antigen-specific VHHs are commonly selected by panning phage libraries on immobilized antigen, e.g., antigen coated onto the plastic surface of a test tube, biotinylated antigens immobilized on streptavidin beads, or membrane proteins expressed on the surface of cells.
- immobilized antigen e.g., antigen coated onto the plastic surface of a test tube
- biotinylated antigens immobilized on streptavidin beads or membrane proteins expressed on the surface of cells.
- VHHs often show lower affinities for their antigen than VHHs derived from animals that have received several immunizations.
- the high affinity of VHHs from immune libraries is attributed to the natural selection of variant VHHs during clonal expansion of B-cells in the lymphoid organs of immunized animals.
- VHHs from non- immune libraries can often be improved by mimicking this strategy in vitro, i.e., by site directed mutagenesis of the CDR regions and further rounds of panning on immobilized antigen under conditions of increased stringency (higher temperature, high or low salt concentration, high or low pH, and low antigen concentrations).
- VHHs derived from camelid are readily expressed in and purified from the E. coli periplasm at much higher levels than the corresponding domains of conventional antibodies.
- VHHs generally display high solubility and stability and can also be readily produced in yeast, plant, and mammalian cells.
- the “Hamers patents” describe methods and techniques for generating VHH against any desired target (see for example US 5,800,988; US 5,874, 541 and US 6,015,695).
- the “Hamers patents” more particularly describe production of VHHs in bacterial hosts such as E. coli (see for example US 6,765,087) and in lower eukaryotic hosts such as moulds (for example Aspergillus or Trichoderma) or in yeast (for example Saccharomyces, Kluyveromyces, Hansenula or Pichia) (see for example US 6,838,254).
- the Tau inhibitor is a peptide, petptidomimetic, small organic molecule, antibody, aptamers, siRNA or antisense oligonucleotide.
- peptidomimetic refers to a small protein-like chain designed to mimic a peptide.
- the Tau inhibitor is an aptamer.
- Aptamers refer to a class of molecule that represents an alternative to antibodies in term of molecular recognition.
- Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.
- Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library, as described in Tuerk C. and Gold L., Science, 1990, 249(4968): 505- 10.
- the random sequence library is obtainable by combinatorial chemical synthesis of DNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence.
- Peptide aptamers consists of a conformationally constrained antibody variable region displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods (Colas et al., Nature, 1996,380, 548-50).
- the Tau inhibitor is a small organic molecule.
- small organic molecule refers to a molecule of a size comparable to those organic molecules generally used in pharmaceuticals.
- Preferred small organic molecules range in size up to about 5000 Da, more preferably up to 2000 Da, and most preferably up to about 1000 Da.
- the Tau inhibitor is a polypeptide.
- a Tau polypeptide may be used as vaccine composition in order to induce an anti Tau serum.
- Tau inhibitors according to the invention is a vaccine composition comprising an isolated peptide of Tau.
- vaccine composition it is herein intended a substance which is able to induce an immune response in an individual, and for example to induce the production of antibodies directed against the isolated tau polypeptide.
- a vaccine is defined herein as a biological agent which is capable of providing a protective response in an animal to which the vaccine has been delivered and is incapable of causing severe disease.
- the vaccine stimulates antibody production or cellular immunity against the pathogen (or agent) causing the disease; administration of the vaccine thus results in immunity from the disease.
- Active immunization with vaccine composition is long lasting because it induces immunological memory. Active vaccines are easy to administer (different routes) and the production is cost-effective. Immunization generates polyclonal response; antibodies can recognize multiple epitopes on the target protein with different affinity and avidity. On the other hand, the immune response depends on the host immune system, there is a variability in the antibody response across patients.
- AADvacl for Alzheimer’s disease and non-fluent primary progressive aphasia (Axon Neuroscience SE)
- ACL35 vaccine for Alzheimer’s disease (AC Immune SA, Janssen).
- Active vaccine AADvacl consists of tau peptide (aa 294-305/4R) that was coupled to keyhole limpet haemocyanin (KLH) in order to stimulate production of specific antibodies.
- ACL35 vaccine is a liposome-based vaccine consisting of a synthetic peptide to mimic the phospho-epitope of tau at residues pS396/pS404 anchored into a lipid bilayer.
- the Tau inhibitor is a PROTAC against Tau.
- PROTACs Protein Engineering Targeting Chimera
- PROTACs Protein Engineering Targeting Chimera
- the PROTAC approach is therefore a chemical protein knock-down strategy. It is therefore could be useful to provide bifunctional chimeric ligands capable of inducing targeted proteolysis of Tau according to the PROTAC strategy.
- Keapl a substrate adaptor protein for ubiquitin E3 ligase involved in oxidative stress regulation, as a novel candidate for PROTACs that can be applied in the degradation of the nonenzymatic protein Tau.
- This peptide PROTAC by recruiting Keapl -Cul3 ubiquitin E3 ligase was developed and applied in the degradation of intracellular Tau.
- Peptide 1 showed strong in vitro binding with Keapl and Tau.
- the Tau inhibitor according to the invention is an inhibitor of Tau gene expression.
- the Tau inhibitor is a short hairpin RNA (shRNA), a small interfering RNA (siRNA) or an antisense oligonucleotide which inhibits the expression of Tau.
- the inhibitor of Tau expression is siRNA
- the inhibitor of Tau expression is shRNA.
- shRNA short hairpin RNA
- RISC RNA-induced silencing complex
- siRNA Small interfering RNA
- siRNA small interfering RNA
- RNAi RNA interference pathway
- Anti-sense oligonucleotides include anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of the targeted mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of the targeted protein, and thus activity, in a cell.
- antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence can be synthesized, e.g., by conventional phosphodiester techniques.
- Methods for using antisense techniques for specifically inhibiting gene expression of genes whose sequence is known are well known in the art (e.g. see U.S. Pat. Nos. 6,566,135; 6,566,131; 6,365,354; 6,410,323; 6,107,091; 6,046,321; and 5,981,732).
- Ribozymes can also function as an inhibitor of Tau expression for use in the present invention.
- Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
- the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
- Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of mineralocorticoid receptor mRNA sequences are thereby useful within the scope of the present invention.
- ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site can be evaluated for predicted structural features, such as secondary structure, that can render the oligonucleotide sequence unsuitable. The suitability of candidate targets can also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using, e.g., ribonuclease protection assays.
- Antisense oligonucleotides, siRNAs, shRNAs of the invention may be delivered in vivo alone or in association with a vector.
- a "vector" is any vehicle capable of facilitating the transfer of the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid to the cells and typically mast cells.
- the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector.
- the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid sequences.
- Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as Moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rous sarcoma virus; adenovirus, adeno-associated virus; S V40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and RNA virus such as a retrovirus.
- retrovirus such as Moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rous sarcoma virus
- adenovirus adeno-associated virus
- S V40-type viruses polyoma viruses
- Epstein-Barr viruses Epstein-Barr viruses
- papilloma viruses herpes virus
- vaccinia virus poli
- Non-cytopathic viral vectors are based on non-cytopathic eukaryotic viruses in which non- essential genes have been replaced with the gene of interest.
- Non-cytopathic viruses include retroviruses (e.g., lentivirus), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA.
- Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replication-deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle).
- retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo.
- viruses for certain applications are the adeno-viruses and adeno-associated viruses, which are double-stranded DNA viruses.
- the adeno-associated virus can be engineered to be replication deficient and is capable of infecting a wide range of cell types and species. It further has advantages such as, heat and lipid solvent stability; high transduction frequencies in cells of diverse lineages, including hemopoietic cells; and lack of superinfection inhibition thus allowing multiple series of transductions.
- the adeno-associated virus can integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection.
- adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event.
- the adeno-associated virus can also function in an extrachromosomal fashion.
- Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g. Sambrook et al., 1989. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigenencoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operatively encoded within the plasmid. These plasmids are well known to those of ordinary skill in the art.
- Plasmids may be custom designed using restriction enzymes and ligation reactions to remove and add specific fragments of DNA. Plasmids may be delivered by a variety of parenteral, mucosal and topical routes.
- the DNA plasmid can be injected by intramuscular, eye, intradermal, subcutaneous, intra-articular or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally. It may also be administered into the epidermis or a mucosal surface using a gene-gun.
- the plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
- the Tau inhibitor is an endonuclease.
- sequencing technologies have provided an unprecedentedly detailed overview of the multiple genetic aberrations in cancer.
- these new data strongly emphasize the need of fast and reliable strategies to characterize the normal and pathological function of these genes and assess their role, in particular as driving factors during oncogenesis.
- the new technologies provide the means to recreate the actual mutations observed in cancer through direct manipulation of the genome. Indeed, natural and engineered nuclease enzymes have attracted considerable attention in the recent years.
- NHEJ error-prone nonhomologous end-joining
- HDR high-fidelity homology-directed repair
- the endonuclease is CRISPR-cas.
- CRISPR-cas has its general meaning in the art and refers to clustered regularly interspaced short palindromic repeats associated which are the segments of prokaryotic DNA containing short repetitions of base sequences.
- the endonuclease is CRISPR-cas9 which is from Streptococcus pyogenes.
- the CRISPR/Cas9 system has been described in US 8697359 Bl and US 2014/0068797. Originally an adaptive immune system in prokaryotes (Barrangou and Marraffini, 2014), CRISPR has been recently engineered into a new powerful tool for genome editing. It has already been successfully used to target important genes in many cell lines and organisms, including human (Mali et al., 2013, Science, Vol. 339 : 823-826), bacteria (Fabre et al., 2014, PLoS Negl. Trop. Dis., Vol.
- the endonuclease is CRISPR-Cpfl which is the more recently characterized CRISPR from Provotella and Francisella 1 (Cpfl) in Zetsche et al. (“Cpfl is a Single RNA-guided Endonuclease of a Class 2 CRISPR-Cas System (2015); Cell; 163, 1-13).
- the Tau inhibitor is an aptamer.
- aptamers are DNA aptamers such as described in Prodeus et al 2015.
- a major disadvantage of aptamers as therapeutic entities is their poor pharmacokinetic profiles, as these short DNA strands are rapidly removed from circulation due to renal filtration.
- aptamers according to the invention are conjugated to with high molecular weight polymers such as polyethylene glycol (PEG).
- PEG polyethylene glycol
- the aptamer is an anti-PD-1 aptamer.
- the anti-PD-1 aptamer is MP7 pegylated as described in Prodeus et al 2015.
- administering refers to the act of injecting or otherwise physically delivering a substance as it exists outside the body (e.g. a Tau inhibitor) into the subject, such as by mucosal, intradermal, intravenous, subcutaneous, intramuscular delivery and/or any other method of physical delivery described herein or known in the art.
- a substance as it exists outside the body (e.g. a Tau inhibitor) into the subject, such as by mucosal, intradermal, intravenous, subcutaneous, intramuscular delivery and/or any other method of physical delivery described herein or known in the art.
- administration of the substance typically occurs after the onset of the disease or symptoms thereof.
- administration of the substance typically occurs before the onset of the disease or symptoms thereof.
- a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result.
- a therapeutically effective amount of drug may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of drug to elicit a desired response in the individual.
- a therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects.
- the efficient dosages and dosage regimens for drug depend on the disease or condition to be treated and may be determined by the persons skilled in the art. A physician having ordinary skill in the art may readily determine and prescribe the effective amount of the pharmaceutical composition required.
- a suitable dose of a composition of the present invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect according to a particular dosage regimen.
- Such an effective dose will generally depend upon the factors described above.
- a therapeutically effective amount for therapeutic use may be measured by its ability to stabilize the progression of disease.
- One of ordinary skill in the art would be able to determine such amounts based on such factors as the subject's size, the severity of the subject's symptoms, and the particular composition or route of administration selected.
- An exemplary, non-limiting range for a therapeutically effective amount of drug is about 0.1- 100 mg/kg, such as about 0.1-50 mg/kg, for example about 0.1-20 mg/kg, such as about 0.1-10 mg/kg, for instance about 0.5, about such as 0.3, about 1, about 3 mg/kg, about 5 mg/kg or about 8 mg/kg.
- Administration may e.g. be intravenous, intramuscular, intraperitoneal, or subcutaneous, and for instance administered proximal to the site of the target. Dosage regimens in the above methods of treatment and uses are adjusted to provide the optimum desired response (e.g., a therapeutic response).
- a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.
- the efficacy of the treatment is monitored during the therapy, e.g. at predefined points in time.
- treatment according to the present invention may be provided as a daily dosage of the agent of the present invention in an amount of about 0.1-100 mg/kg, such as 0.2, 0.5, 0.9, 1.0, 1.1, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 45, 50, 60, 70, 80, 90 or 100 mg/kg, per day, on at least one of days 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40, or alternatively, at least one of weeks 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 after initiation of treatment, or any combination thereof, using single or divided doses every 24, 12, 8, 6, 4, or 2 hours, or any combination thereof.
- 0.1-100 mg/kg such as 0.2, 0.5, 0.9, 1.0, 1.1, 1.5, 2, 3, 4, 5,
- the patient is administered with a pharmaceutical composition comprising the therapeutically effective amount of a Tau inhibitor as active principle and at least one pharmaceutically acceptable excipient.
- the term “active principle” or “active ingredient” are used interchangeably.
- the term “pharmaceutical composition” refers to a composition described herein, or pharmaceutically acceptable salts thereof, with other agents such as carriers and/or excipients.
- the pharmaceutical compositions as provided herewith typically include a pharmaceutically acceptable carrier.
- the term “pharmaceutically acceptable carrier” includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
- Remington's Pharmaceutical-Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutical compositions and known techniques for the preparation thereof.
- a pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
- the pharmaceutical compositions contain vehicles, which are pharmaceutically acceptable for a formulation capable of being injected. These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
- the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
- the form must be sterile and must be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
- Sterile injectable solutions are prepared by incorporating the agent of the present invention in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization.
- dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
- a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
- the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
- FIGURES are a diagrammatic representation of FIGURES.
- Figure 1 Inhibition of Tau increases y -H2AX levels after bleomycin treatment in MCF7 and MDA-MB-231.
- MCF7 and MDA-MB-231 shCtrl or shTau stable clones were treated or not with bleomycin (BLM, 30 pg/ml) for 2h. Cells were then labeled with a y-H2AX antibody to quantify DNA double-strand breaks. Nuclear y-H2AX bulk fluorescence intensity was analyzed using Image J. All experiments were carried out in 3 independent experiments. Data are mean ⁇ SD (n > 60 cells per conditions) **P ⁇ 0.01; ***P ⁇ 0.001..
- Figure 2 Inhibition of Tau increases y -H2AX levels after X-ray treatment in MCF7 and MDA-MB-231.
- MCF7 and B MDA-MB-231 shCtrl or shTau stable clones were X-irradiated with 2 Gy and then further incubated for 5 min, 2, 4 and 6h. Cells were then labeled with a y-H2AX antibody to quantify DNA double-strand breaks and the number of y-H2AX foci per cell was determined using Image J. All experiments were carried out in 3 independent experiments. Data are mean ⁇ SD (n > 60 cells per conditions) * ⁇ 0.05; ** ⁇ 0.01; *** ⁇ 0.001. #P ⁇ 0.05 ### ⁇ 0.001 compared to untreated shCtrl cells. Representative
- FIG. 3 Tau inhibition increases mutation frequency measured by the HPRT test.
- A Western-blot analysis of Tau expression in CHO-GFP and CHO-Tau-GFP stable clones using anti -Tau and anti-actin antibodies
- B Schematic representation of the HPRT test in CHO cells.
- C CHO cells were stably transfected with plasmids coding for GFP or GFP-Tau and treated with cisplatin (10 pM, 2 h) or oxaliplatin (20 pM, 2 h). Mutation frequency was measured as the number of colony/number of cells seeded X efficiency of plating.
- FIG. 4 Tau increases HR and cNHEJ activities.
- A Schematic representation of DR-GFP reporter.
- B Western-blot analyses of a HeLa DR-GFP stable clone transiently transfected with plasmid encoding the endonuclease I-Scel with or without plasmid encoding Tau protein.
- C Tau increases HR activity. HeLa DR-GFP cells were transfected as described in (B). The percentage of GFP-positive cells was quantitated by flow cytometry (five independent experiments). Results are expressed as fold induction relative to the control.
- D Schematic representation of EJ5GFP reporter.
- E Western-blot analyses of HeLa EJ5-GFP stable clone transiently transfected with plasmid encoding the endonuclease I-Scel with or without plasmid encoding Tau protein.
- F Tau increased cNHEJ activity. HeLa EJ5-GFP cells were transfected as described in (E). The percentage of GFP-positive cells was quantitated by flow cytometry (Five independent experiments). Results are expressed as fold induction relative to the control. Data are mean ⁇ SD **PO.OI; ***7’0.001.
- FIG. 5 Tau depletion confers sensitivity to doxorubicin and X-rays in mice xenografts.
- MCF7 cells expressing either scramble (shctrl) or shTau were injected subcutaneously (5 mice per group) and tumor volume was measured for 11 days starting from 100 mm 3 and (A) treated or not with doxorubicin (6 mg/kg x 1) or (B) treated or not with X-rays (2 Gy x 2). Average tumor weight ⁇ SD is shown.
- FIG. 6 Tau increases 53BP1 nuclear translocation after X-ray treatment.
- A MCF7 shCtrl and shTau cells were irradiated with 2 Gy exposure and then further incubated for 15 min. Cells were then labeled with a 53BP1 antibody and the number of 53BP1 foci per cell (n >50) was determined using Image J. All experiments were carried out in 3 independent experiments. Data are mean ⁇ SD 7’0.05; **7’0.01; ***7’0.001.
- B MCF7 shCtrl or shTau stable clones were irradiated with 2 Gy and then further incubated for 15 min.
- FIG. 7 Tau regulates the 53BPl/microtubule interaction.
- A (B) The graph shows the quantification of three independent proximity ligation assays (53BP1-Dynein) experiments as in Proximity Ligation Assay showing 53BP1-Dynein interaction in MCF7shCtrl and shTau cells in control and 2 Gy treated conditions. All data are mean ⁇ SD *7’0.05; **7’0.01; ***7’0.001.
- pimEJ5GFP was a gift from Jeremy Stark (Addgene plasmid # 44026) [25]
- pDRGFP Additional plasmid # 26475
- pCBAScel Additional plasmid # 26477
- Maria Jasin a gift from Maria Jasin [26]
- Short hairpin Tau and RNA Ctrl vectors were purchased from Santacruz.
- Doxorubicin, cisplatin, oxaliplatin, 6-thioguanine and hypoxanthine were purchased from Sigma-Aldrich and bleomycin from Calbiochem.
- Cells were exposed to ionizing radiation (IR) using an X-ray machine (Clinac23X).
- HeLa cells, MCF7, and MDA-MB-231 were cultured in Dulbecco’s Modified Eagle’s Medium with 10% fetal bovine serum, 2 mM L-glutamine and 50 U/ml penicillin/ streptomycin (Gibco) at 37°C in 5% CO2 humidified air.
- CHO-K1 cells were cultured in RPMI1640 (Gibco) with 10% fetal bovine serum, 2 mM L-glutamine and 50 U/ml penicillin/streptomycin (Gibco).
- MCF7 shctrl and shTau cells have been described previously [14], Transient and stable transfection experiments were performed using the lipofectamine LTX reagent according to the manufacturer guideline (Invitrogen). To isolate stably transfected HeLa clones, cells were transfected with the pDRGFP or pimEJ5GFP plasmids and selected with puromycin (2 Dg/ml). Clones were isolated and tested for GFP expression after pCBAScel transfection. Of 6 clones tested for GFP, one was chosen for further studies.
- CHO-K1 GFP or GFP-Tau cells were transfected with plasmids encoding GFP or GFP-Tau and selected with G418 (200 pg/ml). Stably transfected cells were isolated using flow cytometry and cell sorting (Sony SH800).
- Nuclei were recovered by centrifugation for 10 min
- proteins were solubilized in an SDS loading buffer and analyzed by SDS-PAGE.
- Primary antibodies used in Western blotting experiments were directed against Tau Cter [14], 53BP1 (Cell Signaling), H3 (Millipore), Hsp90 (Santa Cruz) and P-actin (Sigma). Secondary antibodies coupled to HRP were from Sigma-Aldrich. Immune complexes were detected using the ECL+ system from Amersham/GE Healthcare and observed with an Image Reader LAS4000 (Fujifilm). Quantification was performed by densitometry using Imaged software.
- MCF7 shCtrl and shTau cells were recovered in buffer A (80 mM Pipes, pH 6.8, 1 mM MgCh, 2 mM EGTA, 30% glycerol, 0.1% Triton X-100, complete protease inhibitors [Roche]). After ultracentrifugation at 100,000 * g at 21 °C for 18 min, supernatants were collected as cytosolic fractions. The pellets (microtubule fraction) were recovered in RIPA buffer and sonicated. Samples were mixed with LDS buffer and equal volumes were loaded for SDS- PAGE and analyzed by immunoblotting.
- Hrprt mutant frequency was determined by measuring the clonogenicity of cells, as previously described [28], Briefly, CHO-K1 cells stably transfected with GFP or GFP-Tau were grown in hypoxanthine medium for 5 days to eliminate preexisting Hprt mutants, then 1.5 x 10 6 cells were plated in 75cm 2 flasks. After 24 h of culture, cells were treated or not with cisplatin (10 pM, 2h), oxaliplatin (20 pM, 2h) or X-rays (2 Gy). Cells were maintained for 8 days to allow the expression of the Hrprt mutant phenotype.
- Proximity ligation assay was performed with a duolink proximity ligation assay kit (Sigma-Aldrich) according to the manufacturer's guidelines. 53BP1 (Millipore) and dynein (Abeam) were used to visualize 53BP1 -dynein proximity.
- HeLa cells with stably integrated HR (DR-GFP) or nonhomologous end joining (EJ5GFP) reporters were plated in 6-well plates and then transfected with a plasmid encoding the LScel endonuclease gene with or without Tau. The cells were left to grow for 48 h and then trypsinized and washed once with PBS. GFP fluorescence was analyzed using an LSR FORTESSA X20 cytometer (Becton Dickinson).
- Tau decreases the mutation rate induced by DNA damaging agents
- Figure 3C and 3D demonstrate that the frequency of spontaneous 6- thioguanine resistant mutants was not significantly different in CHO KI cells expressing Tau compared to GFP control cells.
- GFP or GFP-Tau CHO KI cells were treated with cisplatin (10 pM, 2 h), oxaliplatin (20 pM, 2 h) or X-rays (4 Gy).
- the frequencies of 6- thioguanine induced by these compounds were significantly lower in cells expressing Tau (3.3-, 3.1- and 2.8-fold for cisplatin, oxaliplatin and X-rays respectively).
- these results confirmed a significant role for Tau in the repair of anticancer agent induced-DNA lesions.
- reporter genes of either HR or cNHEJ we employed reporter genes of either HR or cNHEJ.
- HR reporter gene DR-GFP
- DR-GFP the HR reporter gene
- the I-Scel endonuclease generates a DSB.
- Platinum drugs acts mainly by forming intrastrand diadducts, primarily repaired via the nucleotide excision repair system [45], Interestingly, cytosolic sequestration of DNA repair proteins has been linked to neuronal death in different Tauopathies such as Alzheimer’s disease, Pick’s disease, corticobasal neurodegeneration or progressive supranuclear palsy [46,47],
- Tau expression may be of interest as a molecular marker for response to DNA damaging agents and as a beneficial therapeutic target in tumors.
- Microtubule-targeting agents augment the toxicity of DNA-damaging agents by disrupting intracellular trafficking of DNA repair proteins. Proc Natl Acad Set U S A 2015, 112, 1571-1576.
- Giannakakou, P.; Sackett, D.L.; Ward, Y.; Webster, K.R.; Blagosklonny, M.V.; Fojo, T. P53 is associated with cellular microtubules and is transported to the nucleus by dynein. Nat Cell Biol 2000, 2, 709-717.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Zoology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biomedical Technology (AREA)
- Medicinal Chemistry (AREA)
- Molecular Biology (AREA)
- Animal Behavior & Ethology (AREA)
- General Engineering & Computer Science (AREA)
- Immunology (AREA)
- Pharmacology & Pharmacy (AREA)
- Wood Science & Technology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Biotechnology (AREA)
- General Chemical & Material Sciences (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Microbiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Marine Sciences & Fisheries (AREA)
- Gastroenterology & Hepatology (AREA)
- Epidemiology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Therapeutic resistance is one of the major challenges in cancer treatment. These latter may be inherently resistant or this resistance may be acquired during treatment. In this context, the tau protein, encoded by the MAPT gene, could prove to be an important contributor in resistance to cancer therapy. The inventors have been thus abled to confirm in vivo that the tau protein was a factor of resistance to radiotherapy and the chemotherapy inducing DSB (doxorubicin) and that the reduction of its expression by shRNA increased the sensitivity of tumors to these treatments. The present invention relates to a method for decreasing therapeutic acquired resistance to chemotherapy agent and/or radiotherapy agent in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a Tau inhibitor.
Description
METHODS FOR DECREASING THERAPEUTIC ACQUIRED RESISTANCE
TO CHEMOTHERAPY AND/OR RADIOTHERAPY
FIELD OF THE INVENTION:
The present invention is in the field of medicine, in particular oncology, for decreasing therapeutic acquired resistance to chemotherapy and/or radiotherapy in a subject.
BACKGROUND OF THE INVENTION:
Although defects in DNA damage response lead to genomic instability, this can also represent the “Achille’s heel” of cancer cells. Deficiency in any specific repair pathway makes cells more dependent on the remaining DNA-repair pathways, ren-dering them more vulnerable to certain DNA-targeting therapies. Double-strand DNA breaks (DSBs) are considered as the most lethal DNA lesions and are commonly induced in cancer radio- or chemo-therapy. However, one of the main limitations of DSB-associated therapies is the tumor resistance developed before or after treatment [1,2]. Thus, it will be important to decipher the underlying molecular mechanism of this resistance to improve existing regimens.
In cells, DSBs activate DNA damage response pathways through the kinases ATM, ATR and DNA-PKcs, that in turn activate proteins involved in two main pathways, the classical nonhomologous end-joining (cNHEJ) and the homologous recombination (HR). cNHEJ mediates repair by directly rejoining DSB ends, contrary to HR which utilizes a homologous DNA sequence to guide DNA repair [3,4], Which of the two repair routes is used depends in part of the cell cycle phase. Since HR relies on the presence of a sister chromatid, it can be effective only in the late S/G2 phase while the NHEJ pathway is effective throughout the cell cycle. Recruitment of specific factors to DNA lesions is also essential to initiate DNA repair and this is regulated, in part, by histone post-translational modifications [5], In this context, 53BP1 and BRCA1 (breast cancer 1), together with auxiliary factors, play a pivotal role in the pathway choice. They trigger either cNHEJ or HR respectively. 53BP1 blocks DNA end resection necessary for HR. cNHEJ accounts for most DSB repair in mammalian cells [6-8],
DNA repair mechanisms are multifaced but recent studies demonstrated an essential role for microtubules. First, microtubule dynamics was demonstrated to be crucial in the intracellular trafficking of DNA repair proteins [9], Second, it has been demonstrated that the interaction between microtubules and linker of the nucleoskeleton, and cytoskeleton’ complexes increase the mobility of DSBs toward repair complexes [10],
Tau was first described as a neuronal microtubule-associated protein (MAPT) and when it aggregates it is a major player in neurodegenerative diseases like Alzheimer’s [11], However, Tau has many other cellular functions [12], In addition to microtubule dynamics, it interferes with several biological processes such as signaling pathways, RNA metabolism and it even contributes to the inflammatory response. Furthermore, Tau is not only found in the cytoplasm but also in the cell nucleus where it participates in chromatin organisation in neuronal and cancer cells [13,14], Several data suggested a role for Tau in DNA repair [15], In particular, Tau deletion induces an increase in DSBs and a slower DNA repair in neurons after reactive oxygen species induction triggered by hyperthermia [16,17], However, the molecular mechanism remains elusive. In this regard, it has been demonstrated in vitro that Tau protects DNA from free radical damage [18], The current explanation of this protective effect is based on Tau’s ability to bind DNA in a sequence-independent manner [19], Although Tau is known as a neuronal protein, a large body of literature has shown that its expression is increased in several types of cancer and may be associated with acquired resistance to Taxanes [20,21], Tau binds in the same binding site as paclitaxel, and consequently compete with this drug [22],
The role of Tau in genome protection and/or repair suggests that Tau expression in cancer cells could be involved in resistance to conventional anti-cancer treatments, in particular those inducing DNA damage. Starting from this hypothesis, we evaluated the role of Tau in DNA repair in breast cancer cell lines.
SUMMARY OF THE INVENTION:
The present invention is in the field of medicine, in particular oncology, for decreasing therapeutic acquired resistance to chemotherapy and/or radiotherapy in a subject. In particular, the present invention is defined by the claims.
DETAILED DESCRIPTION OF THE INVENTION:
Therapeutic resistance is one of the major challenges in cancer treatment. These latter may be inherently resistant or this resistance may be acquired during treatment. In this context, the tau protein, encoded by the MAPT gene, could prove to be an important contributor in resistance to cancer therapy. Initially described at the neural level, a literature abundant has now demonstrated that the expression of his gene is increased in several types of cancers and could be associated with resistance to taxanes. This effect has been associated with its well- known role regulator of microtubule assembly and disassembly. However, recent work has demonstrated, at the neuronal level, that tau was a pleiotropic protein, involved in several
mechanisms cellular. Among them, its role in genome protection/repair could be directly related to resistance to other anti-cancer treatments, especially those inducing DNA damage.
The inventors show that DSBs from DNA disappear faster after irradiation or bleomycin treatment in MCF7 and MDA-MB-231 cells expressing MAPT in a manner endogenous than in their homologs stably transfected with a tau-directed shRNA (shTau).
This observation has been linked to an increase in the repair activities of the junctiontype DSBs of the non-homologous ends (NHEJ) and homologous recombination (HR). The inventors were able to explain this new role of tau through its ability to increase microtubular trafficking of the 53BP1 protein, a factor essential in the initiation of repair, thus improving its nuclear translocation. Tau has thus a general role in regulating the nuclear trafficking of DNA repair proteins. These results have tested in vivo using a xenograft immunodeficient (SCID) mouse model.
The inventors have been thus abled to confirm in vivo that the tau protein was a factor of resistance to radiotherapy and the chemotherapy either inducing DSB (doxorubicin) or DNA adducts (cisplatin and oxaliplatin) and that the reduction of tau expression by shRNA increased the sensitivity of tumors to these treatments.
In a first embodiment, the present invention relates to a method for decreasing therapeutic acquired resistance to chemotherapy agent and/or radiotherapy agent in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a Tau inhibitor.
In a second embodiment, the present invention relates to a method for preventing and/or treating cancer with acquired resistance to treatment with chemotherapy agent and/or radiotherapy agent in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a Tau inhibitor
As used herein, the term “patient” or “subject” denote a mammal, such as a rodent, a feline, a canine, and a primate. Preferably, a patient according to the invention is a human. Typically, a patient according to the invention refers to any subject (preferably human) afflicted with or susceptible to be afflicted with a cancer.
As used herein, the term "DNA repair" refers to a collection of processes by which a cell identifies and corrects damage to DNA molecules. Single-strand defects are repaired by base excision repair (BER), nucleotide excision repair (NER), or mismatch repair (MMR). Double-strand breaks are repaired by non-homologous end joining (NHEJ), microhomology- mediated end joining (MMEJ), or homologous recombination. After DNA damage, cell cycle checkpoints are activated, which pause the cell cycle to give the cell time to repair the damage
before continuing to divide. Checkpoint mediator proteins include BRCA1, MDC1, 53BP1, p53, ATM, ATR, CHK1, CHK2, and p21.
As used herein, the term "impaired DNA repair" refers to a state in which a mutated cell or a cell with altered gene expression is incapable of DNA repair or has reduced activity of one or more DNA repair pathways or takes longer to repair damage to its DNA as compared to a wild type cell.
As used herein, the terms "cancer" and "tumors" refer to or describe the pathological condition in mammals that is typically characterized by unregulated cell growth. More precisely, in the methods of the invention, diseases, namely tumors that do express/secrete Tau protein are most likely to respond to the Tau inhibitor treatment. In particular, the cancer may be associated with a solid tumor or lymphoma/leukemia (from hematopoietic cell). Examples of cancers that are associated with solid tumor formation include breast cancer, uterine/cervical cancer, oesophageal cancer, pancreatic cancer, colon cancer, colorectal cancer, kidney cancer, ovarian cancer, prostate cancer, head and neck cancer, non small cell lung cancer stomach cancer, tumors of mesenchymal origin (i.e; fibrosarcoma and rhabdomyoscarcoma) thyroid cancer.
More particular examples of such cancers include chronic myeloid leukemia, acute lymphoblastic leukemia, Philadelphia chromosome positive acute lymphoblastic leukemia (Ph+ ALL), squamous cell carcinoma, small-cell lung cancer, non-small cell lung cancer, glioma, gastrointestinal cancer, renal cancer, ovarian cancer, liver cancer, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, stomach cancer, bladder cancer, hepatocarcinoma, breast cancer, colon carcinoma, and head and neck cancer, gastric cancer, germ cell tumor, pediatric sarcoma, multiple myeloma, acute myelogenous leukemia (AML), chronic lymphocytic leukemia, mastocytosis and any symptom associated with mastocytosis.
Various cancers are also encompassed by the scope of the invention, including, but not limited to, the following: carcinoma including that of the bladder (including accelerated and metastatic bladder cancer), breast, colon (including colorectal cancer), kidney, liver, lung (including small and non-small cell lung cancer and lung adenocarcinoma), ovary, prostate, testis, genitourinary tract, lymphatic system, rectum, larynx, pancreas (including exocrine pancreatic carcinoma), esophagus, stomach, gall bladder, cervix, thyroid, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, Tcell lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, hairy cell lymphoma, histiocytic
lymphoma, and Burketts lymphoma; hematopoietic tumors of myeloid lineage including acute and chronic myelogenous leukemias, myelodysplastic syndrome, myeloid leukemia, and promyelocytic, leukemia; tumors of the central and peripheral nervous system including astrocytoma, neuroblastoma, glioma, and schwannomas; tumors of mesenchymal origin including fibrosarcoma, rhabdomyosarcoma, and osteosarcoma; other tumors including melanoma, xenoderma pigmentosum, keratoactanthoma, seminoma, thyroid follicular cancer, and teratocarcinoma; melanoma, unresectable stage III or IV malignant melanoma, squamous cell carcinoma, small-cell lung cancer, non-small cell lung cancer, glioma, gastrointestinal cancer, renal cancer, ovarian cancer, liver cancer, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, stomach cancer, bladder cancer, hepatocarcinoma, breast cancer, colon carcinoma, and head and neck cancer, retinoblastoma, gastric cancer, germ cell tumor, bone cancer, bone tumors, adult malignant fibrous histiocytoma of bone; childhood malignant fibrous histiocytoma of bone, sarcoma, pediatric sarcoma; myelodysplastic syndromes; neuroblastoma; testicular germ cell tumor, intraocular melanoma, myelodysplastic syndromes; myelodysplastic/myeloproliferative diseases, synovial sarcoma.
In a preferred embodiment, the cancer is a hematopoietic cancer, especially a leukemia or lymphoma. In another preferred embodiment of the present invention, the cancer is a solid tumor. For instance, the cancer may be sarcoma and oestosarcoma such as Kaposi sarcome, AIDS-related Kaposi sarcoma, melanoma, in particular ulveal melanoma, and cancers of the head and neck, kidney, ovary, pancreas, prostate, thyroid, lung, esophagus, breast, bladder, colorectum, liver and biliary tract, uterine, appendix, and cervix, testicular cancer, gastrointestinal cancers and endometrial and peritoneal cancers. Preferably the cancer may be sarcoma, melanoma, in particular ulveal melanoma, and cancers of the head and neck, kidney, ovary, pancreas, prostate, thyroid, lung, esophagus, breast, bladder, colorectum, liver, cervix, and endometrial and peritoneal cancers. For instance, the cancer may be selected from the group consisting of breast cancer, hepatocellular carcinoma, colorectal cancer, glioblastoma, melanoma, and head and neck cancer.
In previous study, it was shown that Tau is overexpressed in different human breast, gastric, prostate cancer cell lines and tissues (Gargini et al., 2019). In particular embodiment regarding the method of the present invention, the solid tumor is selected from the group consisting of breast cancer ((Rouzier et al., 2005;Matrone et al., 2010;Spicakova et al., 2010;Li et al., 2013), gastric cancer (Wang Q et al Pathol. Oncol. Res. (2013) 19:429-435), ovarian cancer (Smoter M. et al. Journal of Experimental & Clinical Cancer Research (2013), 32:25) ,
prostate cancer, glioma, Neuroblastoma, Kidney clear cell carcinoma Lung cancer, Pheochromocytoma/Paraganglioma and Neuroendocrine tumors (NETs).
As used herein, the “Double-strand breaks” (DSB) has its general meaning in the art and refers
To the most deleterious types of DNA lesions. DSB processing and repair can cause sequence deletions, loss of heterozygosity, and chromosome rearrangements resulting in cell death or carcinogenesis. There are two major pathways for repairing them: homologous recombination and nonhomologous DNA end joining (NHEJ). The diverse causes of DSBs result in a diverse chemistry of DNA ends that must be repaired. Across NHEJ evolution, the enzymes of the NHEJ pathway exhibit a remarkable degree of structural tolerance in the range of DNA end substrate configurations upon which they can act. In vertebrate cells, the nuclease, polymerases and ligase of NHEJ are the most mechanistically flexible and multifunctional enzymes in each of their classes. Unlike repair pathways for more defined lesions, NHEJ repair enzymes act iteratively, act in any order, and can function independently of one another at each of the two DNA ends being joined. NHEJ is critical not only for the repair of pathologic DSBs as in chromosomal translocations, but also for the repair of physiologic DSBs created during V(D)J recombination and class switch recombination. Therefore, patients lacking normal NHEJ are not only sensitive to ionizing radiation, but also severely immunodeficient
DNA strand breakage can be achieved by ionized radiation (radiotherapy). Radiotherapy includes, but is not limited to, y-rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells. Other radiotherapies include microwaves and UV-irradiation. Other approaches to radiation therapy are also contemplated in the present invention.
The DNA-damaging antitumor agent is preferably selected from the group consisting of an inhibitor of topoisomerases I or II, a DNA crosslinker, a DNA alkylating agent, an anti- metabolic agent and inhibitors of the mitotic spindles.
Inhibitors of topoisomerases I and/or II include, but are not limited to, etoposide, topotecan, camptothecin, irinotecan, amsacrine, intoplicine, anthracyclines such as doxorubicin, epirubicine, daunorubicine, idanrubicine and mitoxantrone. Inhibitors of Topoisomerase I and II include, but are not limited to, intoplecin. In a preferred embodiment, the DNA-damaging antitumor agent is doxorubicin.
DNA crosslinkers include, but are not limited to, cisplatin, carboplatin and oxaliplatin. In a preferred embodiment, the DNA-damaging antitumor agent is selected from the group consisting of carboplatin and oxaliplatin.
Anti-metabolic agents block the enzymes responsible for nucleic acid synthesis or become incorporated into DNA, which produces an incorrect genetic code and leads to apoptosis. Nonexhaustive examples thereof include, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors, and more particularly Methotrexate, Floxuridine, Cytarabine, 6-Mercaptopurine, 6- Thioguanine, Fludarabine phosphate, Pentostatine, 5 -fluorouracil (5-FU), gemcitabine and capecitabine.
The DNA-damaging anti-tumor agent can be alkylating agents including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas, metal salts and triazenes. Nonexhaustive examples thereof include Uracil mustard, Chlormethine, Cyclophosphamide (CYTOXAN(R)), Ifosfamide, Melphalan, Chlorambucil, Pipobroman, Triethylenemelamine, Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Fotemustine, cisplatin, carboplatin, oxaliplatin, thiotepa, Streptozocin, Dacarbazine, and Temozolomide.
Inhibitors of the mitotic spindles include, but are not limited to, , vinorelbine, larotaxel (also called XRP9881; Sanofi-Aventis), XRP6258 (Sanofi-Aventis), BMS-184476 (Bristol- Meyer-Squibb), BMS-188797 (Bristol-Meyer-Squibb), BMS-275183 (Bristol-Meyer-Squibb), ortataxel (also called IDN 5109, BAY 59-8862 or SB-T-101131 ; Bristol-Meyer-Squibb), RPR 109881 A (Bristol-Meyer-20 Squibb), RPR 116258 (Bristol-Meyer-Squibb), NBT-287 (TAPESTRY), Tesetaxel (also called DJ-927), IDN 5390 (INDENA), and MAC-321 (WYETH). Also see the review of Hennenfent & Govindan (2006, Annals of Oncology, 17, 735- 25 749).
Preferably, the DNA-damaging antitumor agent is an inhibitor of topoisomerases I and/or II, a DNA crosslinker, an anti-metabolic agent or a combination thereof. In a preferred embodiment, the DNA damaging antitumor agent is selected from the group consisting of doxorubicin, 5-FU, carboplatin and oxaliplatin or a combination thereof. In a most preferred embodiment, the conjugated DBait is DT01 and the DNA-damaging antitumor agent is selected from the group consisting of doxorubicin, carboplatin, 5-FU and oxaliplatin.
As used herein, the term “taxane” has its general meaning in the art and relates to a class of diterpenes. They were originally identified from plants of the genus Taxus (yews), and feature a taxadiene core. Paclitaxel (Taxol) and docetaxel (Taxotere) are widely used as chemotherapy agents. Cabazitaxel was FDA approved to treat hormone-refractory prostate cancer.
In some embodiment, the DNA-damaging antitumor agent is not a taxane agent. In some embodiment, the DNA-damaging antitumor agent is not the following taxane agent: oxetaxel, paclitaxel, abraxane.
In some embodiment, the taxane is not oxetaxel. In some embodiment, the taxane is not paclitaxel. In some embodiment, the taxane is not abraxane.
In some embodiment, the method of the present invention does not comprise the administration of DNA-damaging antitumor agent does not comprise a taxane agent. In some embodiment, the method of the present invention does not comprise the administration of oxetaxel, paclitaxel, abraxane.
In some embodiment, the DNA-damaging antitumor agent of the present invention does not comprise the taxane-like compounds described in Papin S, Paganetti P. Emerging Evidences for an Implication of the Neurodegeneration -Associated Protein TAU in Cancer. Brain Sci. 2020 Nov 16;10(l 1):862. doi: 10.3390/brainscilOl 10862.
In some embodiment, Tau in cancer cells could be involved in acquired resistance to conventional anti-cancer treatment, in particular those inducing DNA damage. In some embodiment, Tau in cancer cells could be involved in acquired resistance to radiotherapy or to chemotherapy.
As used herein, the term “acquired resistance” indicates that the cancer becomes resistant and/or substantially less response to the effects of the drug after being exposed to it for a certain period of time.
As used herein, the term “radiation therapy” or “radiotherapy” has its general meaning in the art and refers the treatment of cancer with ionizing radiation. Ionizing radiation deposits energy that injures or destroys cells in the area being treated (the target tissue) by damaging their genetic material, making it impossible for these cells to continue to grow. One type of radiation therapy commonly used involves photons, e.g. X-rays. Depending on the amount of energy they possess, the rays can be used to destroy cancer cells on the surface of or deeper in the body. The higher the energy of the x-ray beam, the deeper the x-rays can go into the target tissue. Linear accelerators and betatrons produce x-rays of increasingly greater energy. The use of machines to focus radiation (such as x-rays) on a cancer site is called external beam radiation therapy. Gamma rays are another form of photons used in radiation therapy. Gamma rays are produced spontaneously as certain elements (such as radium, uranium, and cobalt 60) release radiation as they decompose, or decay. In some embodiments, the radiation therapy is external radiation therapy. Examples of external radiation therapy include, but are not limited to, conventional external beam radiation therapy; three-dimensional conformal
radiation therapy (3D-CRT), which delivers shaped beams to closely fit the shape of a tumor from different directions; intensity modulated radiation therapy (IMRT), e.g., helical tomotherapy, which shapes the radiation beams to closely fit the shape of a tumor and also alters the radiation dose according to the shape of the tumor; conformal proton beam radiation therapy; image-guided radiation therapy (IGRT), which combines scanning and radiation technologies to provide real time images of a tumor to guide the radiation treatment; intraoperative radiation therapy (IORT), which delivers radiation directly to a tumor during surgery; stereotactic radiosurgery, which delivers a large, precise radiation dose to a small tumor area in a single session; hyperfractionated radiation therapy, e.g., continuous hyperfractionated accelerated radiation therapy (CHART), in which more than one treatment (fraction) of radiation therapy are given to a subject per day; and hypofractionated radiation therapy, in which larger doses of radiation therapy per fraction is given but fewer fractions.
In some embodiment, the present methods of invention invention relates to radiotherapy inducing double-strans breaks.
Indeed, ionizing radiations cause directly or indirectly double-stranded breaks (DSBs) and trigger cell/tissue death (necrosis or apoptosis). The cytotoxic effect of ionizing radiation forms the basis for radiotherapy, which is widely used in the treatment of human cancer. The efficacy of radiotherapy is currently limited by the radio-resistance of certain tumors (for example, glioblastoma, head and neck squamous cell carcinoma) and by the side effects caused by irradiation of nearby normal tissues (for example, in the treatment of breast and cervical cancer).
As used herein, the term "radiosensitivity" refers to the relative susceptibility of cells to the harmful effect of ionizing radiation. The more radiosensitive a cell is, the less radiation that is required to kill that cell. In general, it has been found that cell radiosensitivity is directly proportional to the rate of cell division and inversely proportional to the cell's capacity for DNA repair.
As used herein, the term "radioresistant" refers to a cell that does not die when exposed to clinically suitable dosages of radiation.
As used herein, the term “radiation-induced toxicity” or “radiotoxicity” designates any toxic or adverse side effect induced by radiation which may be observed in a subject treated by radiation therapy or in an organotypic slice which was irradiated. In particular, the term radiotoxicity includes any toxic or adverse side effect of radiation, which may be observed in the irradiated organotypic slice comprising healthy tissue. The radiotoxicity also includes any
toxic or adverse side effect of radiation, which may be observed in the irradiated organotypic slice comprising healthy tissue and cancer tissue.
As used herein, the term "chemotherapy" has its general meaning in the art and refers to chemical compounds that are effective in inhibiting tumor growth. Examples of chemotherapeutic agents include multikinase inhibitors such as sorafenib and sunitinib, alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, tri ethyl enethiophosphaorarni de and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; cally statin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CBI-TMI); eleutherobin; pancrati statin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estrarnustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimus tine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as the enediyne antibiotics (e.g. calicheamicin, especially calicheamicin (11 and calicheamicin 211, see, e.g., Agnew Chem Inti. Ed. Engl. 33: 183-186 (1994); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, canninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholinodoxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idanrbicin, marcellomycin, mitomycins, mycophenolic acid, nogalarnycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptomgrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5 -fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, 5-FU; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; antiadrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic
acid; aceglatone; aldophospharnide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defo famine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidamol; nitracrine; pento statin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK®; razoxane; rhizoxin; sizofiran; spirogennanium; tenuazonic acid; triaziquone; 2, 2', 2"- trichlorotriethylarnine; trichothecenes (especially T-2 toxin, verracurin A, roridinA and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobromtol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, chlorambucil; gemcitabine; 6- thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisp latin and carbop latin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-11 ; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Also included in this definition are antihormonal agents that act to regulate or inhibit honnone action on tumors such as anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
In some embodiment, the present invention relates to chemotherapy inducing doublestranded breaks (DSBs).
The use of chemotherapeutic agents can cause DNA damages, including direct or indirect double-stranded breaks. Examples of mostly used families of chemotherapeutic agents (chemical cytotoxics) are: inhibitors of topoisomerases I or II (camptothecin/topotecan, epirubicin/etoposide), DNA crosslinkers (cisplatin/carboplatin/oxaliplatin), DNA alkylating agents (carmustine/dacarbazine) or anti-metabolic agents (5- fluorouracil/gemcitabine/capecitabine).
As used herein, the term "chemosensitivity" refers to the relative susceptibility of cancer cells to the effects of anticancer drugs. The more chemosensitive a cancer cell is, the less anticancer drug is required to kill that cell.
As used herein, the term "treatment" or "treat" refer to both prophylactic or preventive treatment as well as curative, improving the patient’s condition or disease modifying treatment, including treatment of patient at risk of contracting the disease or suspected to have contracted
the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse. The treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment. By "therapeutic regimen" is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy. A therapeutic regimen may include an induction regimen and a maintenance regimen. The phrase "induction regimen" or "induction period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease. The general goal of an induction regimen is to provide a high level of drug to a patient during the initial period of a treatment regimen. An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both. The phrase "maintenance regimen" or "maintenance period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a patient during treatment of an illness, e.g., to keep the patient in remission for long periods of time (months or years). A maintenance regimen may employ continuous therapy (e.g., administering a drug at regular intervals, e.g., daily, weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., disease manifestation, etc.]).
As used herein, the term “preventing” intends characterizing a prophylactic method or process that is aimed at delaying or preventing the onset of a disorder or condition to which such term applies.
The term “expression” when used in the context of expression of a gene or nucleic acid refers to the conversion of the information, contained in a gene, into a gene product. A gene product can be the direct transcriptional product of a gene (e.g., mRNA, tRNA, rRNA, antisense RNA, ribozyme, structural RNA or any other type of RNA) or a protein (i.e. Tau) produced by translation of a mRNA.
As used herein, the term “Tau” denotes the Tau protein from mammals and especially from primates (and Tupaiidae). Human Tau is a neuronal microtubule-associated protein found predominantly in axons and functions to promote tubulin polymerization and stabilize microtubules. Six isoforms (isoform A, B, C, D, E, F, G, fetal-Tau) are found in the human
brain, the longest isoform comprising 441 amino acids (isoform F, Uniprot P10636-8). Tau and its properties are also described by Reynolds, C. H. et al., J. Neurochem. 69 (1997) 191-198. Tau, in its hyperphosphorylated form, is the major component of paired helical filaments (PHF), the building block of neurofibrillary lesions in Alzheimer's disease (AD) brain. Tau can be phosphorylated at its serine or threonine residues by several different kinases including GSK3beta, cdk5, MARK and members of the MAP kinase family.
The protein sequence of human Tau protein, and its isoforms, may be found in Uniprot database with the following access numbers:
Tau isoform Fetal (352 Amino Acids) Uniprot Pl 0636-2
Tau isoform B (381 AA) Uniprot Pl 0636-4
Tau isoform D (383 AA) Uniprot P10636-6
Tau isoform C (410 AA) Uniprot P10636-5
Tau isoform E (412 AA) Uniprot Pl 0636-7
Tau isoform F (441 AA) Uniprot P10636-8
Tau isoform G (776 AA) Uniprot Pl 0636-9
In human Tau protein is encoded by the MAPT (Microtubule associated protein tau) gene located on chromosome 17 (Gene ID: 4137). This gene has 18 transcripts (splice variants), 1 gene allele, 255 orthologues, 1 paralogue and is associated with 13 phenotypes.
Example of human MAPT transcripts which encoded Tau protein may be found in Ensembl database with the following access number
Transcript MAPT-201 (833 AA) Ensembl ID ENST00000262410 (Protein coding)
Transcript MAPT-202 (352 AA) Ensembl ID ENST00000334239 (Protein coding Transcript MAPT-203(736 AA) Ensembl ID ENST00000344290 (Protein coding) Transcript MAPT-204 (441 AA) Ensembl ID ENST00000351559 (Protein coding) Transcript MAPT-205 (776 AA) Ensembl ID ENST00000415613 (Protein coding) Transcript MAPT-206 (412 AA) Ensembl ID :ENST00000420682 (Protein coding) Transcript MAPT-207 (410 AA) Ensembl ID ENST00000431008 (Protein coding) Transcript MAPT-208 (383 AA) Ensembl ID ENST00000446361 (Protein coding)
Transcipt MAPT-209 (381 AA) Ensembl ID ENST00000535772 (Protein coding) Transcript MAPT-212 (758 AA) Ensembl ID ENST00000571987 (Protein coding) Transript MAPT-214 (441 AA) Ensembl ID ENST00000574436 (Protein coding) Transcript MAPT-217 (412 AA) Ensembl ID ENST00000680542 (Protein coding) Transcript MAPT-218 (424 AA) Ensembl ID ENST00000680674 (Protein coding)
Of course variant sequences of the Tau may be used in the context of the present invention, those including but not limited to functional homologues, paralogues or orthologues of such sequences.
The term “Tau” should be understood broadly, it encompasses the native Tau, variants thereof having binding activity with microtubule and fragments thereof having binding activity with microtubule. In particular the native Tau, variants and isoforms preferably contain at least three or four microtubule binding domains (named 3R and 4R respectively). All human Tau isoform and MAPT transcript above described contains at least three or four microtubule binding domains.
In a particular embodiment, the Tau protein used in the context of the present invention is transcript Variant (or Tau Isoform) selected from the list consisting of (1N4R) Transcript MAPT-206 (412 AA) (Protein coding Tau isoform E) and (2N4R) Transript MAPT-214 (441 AA) (Protein coding Tau isoform F).
• Tau Inhibitor
In some embodiment, the inhibitor of the present invention is Tau inhibitor.
As used herein, the term "inhibitor" as used herein includes not only drugs for inhibiting activity of target molecules, but also drugs for inhibiting the expression of target molecules.
As used herein, the term “Tau inhibitor” denotes a molecule or compound which can inhibit directly or indirectly the activity of the protein by limiting or impairing the interactions of the protein (ie with microtubules associated with translocation of DNA repair factors such as 53BP1 or with chromatin proteins and DNA), or a molecule or compound which destabilizes the protein structure, or a molecule or compound which inhibits the transcription or the translation of Tau, or accelerates its degradation. The term “Tau inhibitor” also denotes an inhibitor of the expression of the gene coding for the protein.
A specific embodiment, the Tau inhibitor is a Tau inhibitor which directly binds to tau (protein or nucleic sequence (DNA or mRNA)) and neutralizes, blocks, inhibits, abrogates, reduces or interferes with the biological activity of Tau protein such as 1) Tau protein binding activity with microtubules (associated with translocation of DNA repair factors such as 53BP1) and/or 2) initiation of repair activities of the junction-type DSBs of the non-homologous ends (NHEJ) and homologous recombination (HR) .
Accordingly, in the context of the present invention, the Tau inhibitor (i) directly binds to Tau (protein or nucleic sequence (DNA or mRNA)) and (ii) inhibits biological activity of Tau protein such as Tau protein binding activity with microtubules (associated with translocation in nucleus of DNA repair factors such as 53BP1) and/or initiation of repair
activities of the junction-type DSBs of the non-homologous ends (NHEJ) and homologous recombination (HR). Examples of Tau inhibitors include but are not limited to any of the inhibitors described in “Jadhav et al. Acta Neuropathologica Communications (2019) 7:22 all of which are herein incorporated by reference.
By "biological activity" of Tau protein is meant in the context of the present invention, the translocation in nucleus of DNA repair factors such as 53BP1) and/or initiation of repair activities of the junction-type DSBs of the non-homologous ends (NHEJ) and homologous recombination (HR)
Tests for determining the capacity of a compound to be a Tau protein inhibitor are well known to the person skilled in the art. In a preferred embodiment, the antagonist/inhibitor specifically binds to Tau protein (protein or nucleic sequence (DNA or mRNA)) in a sufficient manner to inhibit the biological activity of Tau protein. Binding to Tau protein and inhibition of the biological activity of Tau protein may be determined by any competing assays well known in the art. For example, the assay may consist in determining the ability of the agent to be tested as a Tau protein inhibitor to bind to Tau protein. The binding ability is reflected by the Kd measurement. The term "Kd", as used herein, is intended to refer to the dissociation constant, which is obtained from the ratio of Kd to Ka (i.e. Kd/Ka) and is expressed as a molar concentration (M). Kd values for binding biomolecules can be determined using methods well established in the art. In specific embodiments, an antagonist / inhibitor that "specifically binds to Tau protein" is intended to refer to an inhibitor that binds to human Tau protein polypeptide with a Kd of IpM or less, lOOnM or less, lOnM or less, or 3nM or less. Then a competitive assay may be settled to determine the ability of the agent to inhibit biological activity of Tau protein : inhibition of Tau protein binding with microtubules (associated with translocation in nucleus of DNA repair factors such as 53BP1) and/or initiation of repair activities of the junction-type DSBs of the non-homologous ends (NHEJ) and homologous recombination (HR).
By “inhibitor of the Tau protein activity”, it is herein referred to a compound which is capable of reducing or suppressing 1) the translocation in nucleus of DNA repair factors such as 53BP1 and/or 2) initiation of repair activities of the junction-type DSBs of the non- homologous ends (NHEJ) and homologous recombination (HR).
In view of the teaching of the present disclosure, particularly of the examples, it falls within the ability of the skilled person to assess whether a compound is an inhibitor 1) the translocation in nucleus of DNA repair factors such as 53BP1 and/or 2) initiation of repair activities of the junction-type DSBs of the non-homologous ends (NHEJ) and homologous recombination (HR). A suitable test for detecting Tau protein binding activity with
microtubules (associated with translocation in nucleus of DNA repair factors such as 53BP1) is described in examples hereinafter (see figure 6 and 7).
A suitable test for detecting repair activities of the junction-type DSBs of the non- homologous ends (NHEJ) is described in examples hereinafter (see figure 1 to 4) . DSBs is quantified using a phosphorylated form of H2AX as an indicator of DNA damagefsee 29],
Typically, a tau inhibitor according to the invention includes but is not limited to:
A) Inhibitor of Tau activity selected from the list consisting Anti-Tau antibody and anti- Tau aptamers, tau peptide (vaccines)
B) PROTAC (“Proteolysis Targeting Chimera”) which simultaneously bind a tau protein and an E3 -ubiquitin ligase.
C) Inhibitor of Tau gene expression selected from the list consisting of antisense, oligonucleotide, nuclease, siRNA, shRNA or ribozyme nucleic acid sequence.
In some embodiments, the Tau inhibitor is an antibody.
As used herein, the term “antibody” is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, multi-specific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity. The term includes antibody fragments that comprise an antigen binding domain such as Fab', Fab, F(ab')2, single domain antibodies (DABs), TandAbs dimer, Fv, scFv (single chain Fv), dsFv, ds-scFv, Fd, linear antibodies, VHH (antigen binding fragment of heavy chain only antibodies), minibodies, diabodies, bi specific antibody fragments, bibody, tribody (scFv-Fab fusions, bispecific or trispecific, respectively); sc-diabody; kappa(lamda) bodies (scFv-CL fusions); BiTE (Bispecific T-cell Engager, scFv-scFv tandems to attract T cells); DVD-Ig (dual variable domain antibody, bispecific format); SIP (small immunoprotein, a kind of minibody); SMIP ("small modular immunopharmaceutical" scFv-Fc dimer; DART (ds-stabilized diabody "Dual Affinity ReTargeting"); small antibody mimetics comprising one or more CDRs and the like. The techniques for preparing and using various antibody-based constructs and fragments are well known in the art (see Kabat et al., 1991, specifically incorporated herein by reference). Diabodies, in particular, are further described in EP 404, 097 and WO 93/1 1 161; whereas linear antibodies are further described in Zapata et al. (1995). Antibodies can be fragmented using conventional techniques. For example, F(ab')2 fragments can be generated by treating the antibody with pepsin. The resulting F(ab')2 fragment can be treated to reduce disulfide bridges to produce Fab' fragments. Papain digestion can lead to the formation of Fab fragments. Fab, Fab' and F(ab')2, scFv, Fv, dsFv, Fd, dAbs, TandAbs, ds-scFv, dimers, minibodies, diabodies, bispecific antibody fragments and other fragments can
also be synthesized by recombinant techniques or can be chemically synthesized. Techniques for producing antibody fragments are well known and described in the art. For example, each of Beckman et al., 2006; Holliger & Hudson, 2005; Le Gall et al., 2004; Reff & Heard, 2001 ; Reiter et al., 1996; and Young et al., 1995 further describe and enable the production of effective antibody fragments. In some embodiments, the antibody is a “chimeric” antibody as described in U.S. Pat. No. 4,816,567. In some embodiments, the antibody is a humanized antibody, such as described U.S. Pat. Nos. 6,982,321 and 7,087,409. In some embodiments, the antibody is a human antibody. A “human antibody” such as described in US 6,075,181 and 6,150,584. In some embodiments, the antibody is a single domain antibody such as described in EP 0 368 684, WO 06/030220 and WO 06/003388. In a particular embodiment, the inhibitor is a monoclonal antibody. Monoclonal antibodies can be prepared and isolated using any technique to produce antibody molecules by continuous cell lines in culture. Techniques for production and isolation include but are not limited to the hybridoma technique, the human B-cell hybridoma technique and the EBV-hybridoma technique. In a particular embodiment, the antibody is specific of the isoform B of Tau.
Antibodies directed against Tau can be raised according to known methods by administering the appropriate antigen or epitope to a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others. Various adjuvants known in the art can be used to enhance antibody production. Although antibodies useful in practicing the invention can be polyclonal, monoclonal antibodies are preferred. Monoclonal antibodies against TAU can be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture. Techniques for production and isolation include but are not limited to the hybridoma technique originally described by Kohler and Milstein (1975); the human B-cell hybridoma technique (Cote et al., 1983); and the EBV-hybridoma technique (Cole et al. 1985). Alternatively, techniques described for the production of single chain antibodies (see e.g., U.S. Pat. No. 4,946,778) can be adapted to produce anti-TAU single chain antibodies. Compounds useful in practicing the present invention also include anti-TAU antibody fragments including but not limited to F(ab')2 fragments, which can be generated by pepsin digestion of an intact antibody molecule, and Fab fragments, which can be generated by reducing the disulfide bridges of the F(ab')2 fragments. Alternatively, Fab and/or scFv expression libraries can be constructed to allow rapid identification of fragments having the desired specificity to Tau. Humanized anti-TAU antibodies and antibody fragments therefrom can also be prepared according to known techniques. "Humanized antibodies" are forms of nonhuman (e.g., rodent) chimeric antibodies that contain minimal sequence derived from non-
human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (CDRs) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity. In some instances, framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. Methods for making humanized antibodies are described, for example, by Winter (U.S. Pat. No. 5,225,539) and Boss (Celltech, U.S. Pat. No. 4,816,397).
Tau inhibitors such as anti Tau antibodies are well known in the art. Examples of patents disclosing anti Tau antibodies are. WO/2012/049570, WO/2014096321, WO/2015/004163; WO/2015200806, WO/2016/112078, WO/2018/152359, WO/2020/ 120644 (VHH anti Tau) WO/2020193520, WO/2021/010712,. . .
In the context of the invention, it could be advantageous to use a nanobody directed against Tau in order to enter the cell. Thus in another embodiment, the antibody according to the invention is a single domain antibody directed against Tau. The term “single domain antibody” (sdAb) or "VHH" refers to the single heavy chain variable domain of antibodies of the type that can be found in Camelid mammals which are naturally devoid of light chains. Such VHH are also called “nanobody®”. According to the invention, sdAb can particularly be llama sdAb. The term “VHH” refers to the single heavy chain having 3 complementarity determining regions (CDRs): CDR1, CDR2 and CDR3. The term “complementarity determining region” or “CDR” refers to the hypervariable amino acid sequences which define the binding affinity and specificity of the VHH. The VHH according to the invention can readily be prepared by an ordinarily skilled artisan using routine experimentation. The VHH variants and modified form thereof may be produced under any known technique in the art such as in-vitro maturation. VHHs or sdAbs are usually generated by PCR cloning of the V-domain repertoire from blood, lymph node, or spleen cDNA obtained from immunized animals into a phage display vector, such as pHEN2. Antigen-specific VHHs are commonly selected by panning phage libraries on
immobilized antigen, e.g., antigen coated onto the plastic surface of a test tube, biotinylated antigens immobilized on streptavidin beads, or membrane proteins expressed on the surface of cells. However, such VHHs often show lower affinities for their antigen than VHHs derived from animals that have received several immunizations. The high affinity of VHHs from immune libraries is attributed to the natural selection of variant VHHs during clonal expansion of B-cells in the lymphoid organs of immunized animals. The affinity of VHHs from non- immune libraries can often be improved by mimicking this strategy in vitro, i.e., by site directed mutagenesis of the CDR regions and further rounds of panning on immobilized antigen under conditions of increased stringency (higher temperature, high or low salt concentration, high or low pH, and low antigen concentrations). VHHs derived from camelid are readily expressed in and purified from the E. coli periplasm at much higher levels than the corresponding domains of conventional antibodies. VHHs generally display high solubility and stability and can also be readily produced in yeast, plant, and mammalian cells. For example, the “Hamers patents” describe methods and techniques for generating VHH against any desired target (see for example US 5,800,988; US 5,874, 541 and US 6,015,695). The “Hamers patents” more particularly describe production of VHHs in bacterial hosts such as E. coli (see for example US 6,765,087) and in lower eukaryotic hosts such as moulds (for example Aspergillus or Trichoderma) or in yeast (for example Saccharomyces, Kluyveromyces, Hansenula or Pichia) (see for example US 6,838,254).
Examples of patent disclosing VHH anti Tau antibodies is. WO/2020/120644
In a particular embodiment, the Tau inhibitor is a peptide, petptidomimetic, small organic molecule, antibody, aptamers, siRNA or antisense oligonucleotide. The term “peptidomimetic” refers to a small protein-like chain designed to mimic a peptide.
In a particular embodiment, the Tau inhibitor is an aptamer.
The term “Aptamers” refer to a class of molecule that represents an alternative to antibodies in term of molecular recognition. Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity. Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library, as described in Tuerk C. and Gold L., Science, 1990, 249(4968): 505- 10. The random sequence library is obtainable by combinatorial chemical synthesis of DNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence. Possible modifications, uses and advantages of this class of molecules have been reviewed in Jayasena S.D., Clin. Chem., 1999, 45(9): 1628-50. Peptide aptamers consists of a conformationally constrained antibody variable
region displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods (Colas et al., Nature, 1996,380, 548-50).
In a particular embodiment, the Tau inhibitor is a small organic molecule.
The term “small organic molecule” refers to a molecule of a size comparable to those organic molecules generally used in pharmaceuticals. The term excludes biological macromolecules (e.g., proteins, nucleic acids, etc.). Preferred small organic molecules range in size up to about 5000 Da, more preferably up to 2000 Da, and most preferably up to about 1000 Da.
In some embodiment, the Tau inhibitor is a polypeptide. In a particular embodiment a Tau polypeptide may be used as vaccine composition in order to induce an anti Tau serum.
Accordingly, another example of Tau inhibitors according to the invention is a vaccine composition comprising an isolated peptide of Tau.
By “vaccine composition” it is herein intended a substance which is able to induce an immune response in an individual, and for example to induce the production of antibodies directed against the isolated tau polypeptide.
A vaccine is defined herein as a biological agent which is capable of providing a protective response in an animal to which the vaccine has been delivered and is incapable of causing severe disease. The vaccine stimulates antibody production or cellular immunity against the pathogen (or agent) causing the disease; administration of the vaccine thus results in immunity from the disease. Active immunization with vaccine composition is long lasting because it induces immunological memory. Active vaccines are easy to administer (different routes) and the production is cost-effective. Immunization generates polyclonal response; antibodies can recognize multiple epitopes on the target protein with different affinity and avidity. On the other hand, the immune response depends on the host immune system, there is a variability in the antibody response across patients. Like their passive immunotherapy counterparts, active vaccines targeting the mid-region, microtubule binding domain of Tau and C -terminus of Tau have been extensively investigated in preclinical studies (see table 3 of Jadhav et al. Acta Neuropathologica Communications (2019) 7:22). There are two tau active vaccines that have been tested in human clinical trials, AADvacl for Alzheimer’s disease and non-fluent primary progressive aphasia (Axon Neuroscience SE), and ACL35 vaccine for Alzheimer’s disease (AC Immune SA, Janssen). Active vaccine AADvacl consists of tau peptide (aa 294-305/4R) that was coupled to keyhole limpet haemocyanin (KLH) in order to stimulate production of specific antibodies. ACL35 vaccine is a liposome-based vaccine
consisting of a synthetic peptide to mimic the phospho-epitope of tau at residues pS396/pS404 anchored into a lipid bilayer.
In some embodiment, the Tau inhibitor is a PROTAC against Tau.
As used herein, the term “PROTACs” (“Proteolysis Targeting Chimera”) means bifunctional molecules which simultaneously bind a target protein and an E3-ubiquitin ligase. This causes the poly-ubiquitination of the target protein which is thus degraded into small peptides and amino acids by the proteasome complex. The PROTAC approach is therefore a chemical protein knock-down strategy. It is therefore could be useful to provide bifunctional chimeric ligands capable of inducing targeted proteolysis of Tau according to the PROTAC strategy.
An example of PROTAC targeting Tau is describe Lu M. et al “Discovery of a Keapl- dependent peptide PROTAC to knockdown Tau by ubiquitination-proteasome degradation pathway European Journal of Medicinal Chemistry 146 (2018) 251e259. Briefly in this study authors identified Keapl, a substrate adaptor protein for ubiquitin E3 ligase involved in oxidative stress regulation, as a novel candidate for PROTACs that can be applied in the degradation of the nonenzymatic protein Tau. This peptide PROTAC by recruiting Keapl -Cul3 ubiquitin E3 ligase was developed and applied in the degradation of intracellular Tau. Peptide 1 showed strong in vitro binding with Keapl and Tau.
In another embodiment, the Tau inhibitor according to the invention is an inhibitor of Tau gene expression. In some embodiments, the Tau inhibitor is a short hairpin RNA (shRNA), a small interfering RNA (siRNA) or an antisense oligonucleotide which inhibits the expression of Tau.
In a particular embodiment, the inhibitor of Tau expression is siRNA
In a particular embodiment, the inhibitor of Tau expression is shRNA.
A short hairpin RNA (shRNA) is a sequence of RNA that makes a tight hairpin turn that can be used to silence gene expression via RNA interference. shRNA is generally expressed using a vector introduced into cells, wherein the vector utilizes the U6 promoter to ensure that the shRNA is always expressed. This vector is usually passed on to daughter cells, allowing the gene silencing to be inherited. The shRNA hairpin structure is cleaved by the cellular machinery into siRNA, which is then bound to the RNA-induced silencing complex (RISC). This complex binds to and cleaves mRNAs that match the siRNA to which it is bound.
Small interfering RNA (siRNA), sometimes known as short interfering RNA or silencing RNA, are a class of 20-25 nucleotide-long double- stranded RNA molecules that play a variety of roles in biology. Most notably, siRNA is involved in the RNA interference (RNAi)
pathway whereby the siRNA interferes with the expression of a specific gene. Anti-sense oligonucleotides include anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of the targeted mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of the targeted protein, and thus activity, in a cell. For example, antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence can be synthesized, e.g., by conventional phosphodiester techniques. Methods for using antisense techniques for specifically inhibiting gene expression of genes whose sequence is known are well known in the art (e.g. see U.S. Pat. Nos. 6,566,135; 6,566,131; 6,365,354; 6,410,323; 6,107,091; 6,046,321; and 5,981,732).
Ribozymes can also function as an inhibitor of Tau expression for use in the present invention. Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. The mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage. Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of mineralocorticoid receptor mRNA sequences are thereby useful within the scope of the present invention. Specific ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site can be evaluated for predicted structural features, such as secondary structure, that can render the oligonucleotide sequence unsuitable. The suitability of candidate targets can also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using, e.g., ribonuclease protection assays.
Antisense oligonucleotides, siRNAs, shRNAs of the invention may be delivered in vivo alone or in association with a vector. In its broadest sense, a "vector" is any vehicle capable of facilitating the transfer of the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid to the cells and typically mast cells. Typically, the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector. In general, the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid sequences. Viral vectors are a preferred type of vector and
include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as Moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rous sarcoma virus; adenovirus, adeno-associated virus; S V40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and RNA virus such as a retrovirus. One can readily employ other vectors not named but known to the art.
Preferred viral vectors are based on non-cytopathic eukaryotic viruses in which non- essential genes have been replaced with the gene of interest. Non-cytopathic viruses include retroviruses (e.g., lentivirus), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA. Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replication-deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle). Such genetically altered retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo. Standard protocols for producing replication-deficient retroviruses (including the steps of incorporation of exogenous genetic material into a plasmid, transfection of a packaging cell lined with plasmid, production of recombinant retroviruses by the packaging cell line, collection of viral particles from tissue culture media, and infection of the target cells with viral particles) are provided in Kriegler, 1990 and in Murry, 1991.
Preferred viruses for certain applications are the adeno-viruses and adeno-associated viruses, which are double-stranded DNA viruses. The adeno-associated virus can be engineered to be replication deficient and is capable of infecting a wide range of cell types and species. It further has advantages such as, heat and lipid solvent stability; high transduction frequencies in cells of diverse lineages, including hemopoietic cells; and lack of superinfection inhibition thus allowing multiple series of transductions. Reportedly, the adeno-associated virus can integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection. In addition, wild-type adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event. The adeno-associated virus can also function in an extrachromosomal fashion.
Other vectors include plasmid vectors. Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g. Sambrook et al., 1989. In the
last few years, plasmid vectors have been used as DNA vaccines for delivering antigenencoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operatively encoded within the plasmid. These plasmids are well known to those of ordinary skill in the art. Additionally, plasmids may be custom designed using restriction enzymes and ligation reactions to remove and add specific fragments of DNA. Plasmids may be delivered by a variety of parenteral, mucosal and topical routes. For example, the DNA plasmid can be injected by intramuscular, eye, intradermal, subcutaneous, intra-articular or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally. It may also be administered into the epidermis or a mucosal surface using a gene-gun. The plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
In some embodiments, the Tau inhibitor is an endonuclease. In the last few years, staggering advances in sequencing technologies have provided an unprecedentedly detailed overview of the multiple genetic aberrations in cancer. By considerably expanding the list of new potential oncogenes and tumor suppressor genes, these new data strongly emphasize the need of fast and reliable strategies to characterize the normal and pathological function of these genes and assess their role, in particular as driving factors during oncogenesis. As an alternative to more conventional approaches, such as cDNA overexpression or downregulation by RNA interference, the new technologies provide the means to recreate the actual mutations observed in cancer through direct manipulation of the genome. Indeed, natural and engineered nuclease enzymes have attracted considerable attention in the recent years. The mechanism behind endonuclease-based genome inactivating generally requires a first step of DNA single or double strand break, which can then trigger two distinct cellular mechanisms for DNA repair, which can be exploited for DNA inactivating: the error-prone nonhomologous end-joining (NHEJ) and the high-fidelity homology-directed repair (HDR).
In a particular embodiment, the endonuclease is CRISPR-cas.
As used herein, the term “CRISPR-cas” has its general meaning in the art and refers to clustered regularly interspaced short palindromic repeats associated which are the segments of prokaryotic DNA containing short repetitions of base sequences.
In some embodiment, the endonuclease is CRISPR-cas9 which is from Streptococcus pyogenes. The CRISPR/Cas9 system has been described in US 8697359 Bl and US
2014/0068797. Originally an adaptive immune system in prokaryotes (Barrangou and Marraffini, 2014), CRISPR has been recently engineered into a new powerful tool for genome editing. It has already been successfully used to target important genes in many cell lines and organisms, including human (Mali et al., 2013, Science, Vol. 339 : 823-826), bacteria (Fabre et al., 2014, PLoS Negl. Trop. Dis., Vol. 8:e2671.), zebrafish (Hwang et al., 2013, PLoS One, Vol. 8:e68708.), C. elegans (Hai et al., 2014 Cell Res. doi: 10.1038/cr.2014.11.), bacteria (Fabre et al., 2014, PLoS Negl. Trop. Dis., Vol. 8:e2671.), plants (Mali et al., 2013, Science, Vol. 339 : 823-826), Xenopus tropicalis (Guo et al., 2014, Development, Vol. 141 : 707-714.), yeast (DiCarlo et al., 2013, Nucleic Acids Res., Vol. 41 : 4336-4343.), Drosophila (Gratz et al., 2014 Genetics, doi: 10.1534/genetics.113.160713), monkeys (Niu et al., 2014, Cell, Vol. 156 : 836- 843.), rabbits (Yang et al., 2014, J. Mol. Cell Biol., Vol. 6 : 97-99.), pigs (Hai et al., 2014, Cell Res. doi: 10.1038/cr.2014.11.), rats (Ma et al., 2014, Cell Res., Vol. 24 : 122-125.) and mice (Mashiko et al., 2014, Dev. Growth Differ. Vol. 56 : 122-129.). Several groups have now taken advantage of this method to introduce single point mutations (deletions or insertions) in a particular target gene, via a single gRNA. Using a pair of gRNA-directed Cas9 nucleases instead, it is also possible to induce large deletions or genomic rearrangements, such as inversions or translocations. A recent exciting development is the use of the dCas9 version of the CRISPR/Cas9 system to target protein domains for transcriptional regulation, epigenetic modification, and microscopic visualization of specific genome loci.
In some embodiment, the endonuclease is CRISPR-Cpfl which is the more recently characterized CRISPR from Provotella and Francisella 1 (Cpfl) in Zetsche et al. (“Cpfl is a Single RNA-guided Endonuclease of a Class 2 CRISPR-Cas System (2015); Cell; 163, 1-13).
In some embodiments, the Tau inhibitor is an aptamer.
In a particular embodiment, aptamers are DNA aptamers such as described in Prodeus et al 2015. A major disadvantage of aptamers as therapeutic entities is their poor pharmacokinetic profiles, as these short DNA strands are rapidly removed from circulation due to renal filtration. Thus, aptamers according to the invention are conjugated to with high molecular weight polymers such as polyethylene glycol (PEG). In a particular embodiment, the aptamer is an anti-PD-1 aptamer. Particularly, the anti-PD-1 aptamer is MP7 pegylated as described in Prodeus et al 2015.
As used herein the terms "administering" or "administration" refer to the act of injecting or otherwise physically delivering a substance as it exists outside the body (e.g. a Tau inhibitor) into the subject, such as by mucosal, intradermal, intravenous, subcutaneous, intramuscular delivery and/or any other method of physical delivery described herein or known
in the art. When a disease, or a symptom thereof, is being treated, administration of the substance typically occurs after the onset of the disease or symptoms thereof. When a disease or symptoms thereof, are being prevented, administration of the substance typically occurs before the onset of the disease or symptoms thereof.
A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result. A therapeutically effective amount of drug may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of drug to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects. The efficient dosages and dosage regimens for drug depend on the disease or condition to be treated and may be determined by the persons skilled in the art. A physician having ordinary skill in the art may readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician could start doses of drug employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. In general, a suitable dose of a composition of the present invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect according to a particular dosage regimen. Such an effective dose will generally depend upon the factors described above. For example, a therapeutically effective amount for therapeutic use may be measured by its ability to stabilize the progression of disease. One of ordinary skill in the art would be able to determine such amounts based on such factors as the subject's size, the severity of the subject's symptoms, and the particular composition or route of administration selected. An exemplary, non-limiting range for a therapeutically effective amount of drug is about 0.1- 100 mg/kg, such as about 0.1-50 mg/kg, for example about 0.1-20 mg/kg, such as about 0.1-10 mg/kg, for instance about 0.5, about such as 0.3, about 1, about 3 mg/kg, about 5 mg/kg or about 8 mg/kg. Administration may e.g. be intravenous, intramuscular, intraperitoneal, or subcutaneous, and for instance administered proximal to the site of the target. Dosage regimens in the above methods of treatment and uses are adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. In some embodiments, the efficacy of the treatment is monitored during the therapy, e.g. at predefined points in time. As non-limiting examples, treatment according to the present invention may be provided as a
daily dosage of the agent of the present invention in an amount of about 0.1-100 mg/kg, such as 0.2, 0.5, 0.9, 1.0, 1.1, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 45, 50, 60, 70, 80, 90 or 100 mg/kg, per day, on at least one of days 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40, or alternatively, at least one of weeks 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 after initiation of treatment, or any combination thereof, using single or divided doses every 24, 12, 8, 6, 4, or 2 hours, or any combination thereof.
In some embodiments, the patient is administered with a pharmaceutical composition comprising the therapeutically effective amount of a Tau inhibitor as active principle and at least one pharmaceutically acceptable excipient.
As used herein the term “active principle” or “active ingredient” are used interchangeably. As used herein, the term “pharmaceutical composition” refers to a composition described herein, or pharmaceutically acceptable salts thereof, with other agents such as carriers and/or excipients. The pharmaceutical compositions as provided herewith typically include a pharmaceutically acceptable carrier.
As used herein, the term “pharmaceutically acceptable carrier” includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Remington's Pharmaceutical-Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutical compositions and known techniques for the preparation thereof.
A pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. Typically, the pharmaceutical compositions contain vehicles, which are pharmaceutically acceptable for a formulation capable of being injected. These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the
extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. Sterile injectable solutions are prepared by incorporating the agent of the present invention in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.
FIGURES:
Figure 1 : Inhibition of Tau increases y -H2AX levels after bleomycin treatment in MCF7 and MDA-MB-231. (A) MCF7 and (B) MDA-MB-231 shCtrl or shTau stable clones were treated or not with bleomycin (BLM, 30 pg/ml) for 2h. Cells were then labeled with a y-H2AX antibody to quantify DNA double-strand breaks. Nuclear y-H2AX bulk fluorescence intensity was analyzed using Image J. All experiments were carried out in 3 independent experiments. Data are mean ± SD (n > 60 cells per conditions) **P<0.01; ***P<0.001..
Figure 2: Inhibition of Tau increases y -H2AX levels after X-ray treatment in MCF7 and MDA-MB-231. (A) MCF7 and (B) MDA-MB-231 shCtrl or shTau stable clones were X-irradiated with 2 Gy and then further incubated for 5 min, 2, 4 and 6h. Cells were then labeled with a y-H2AX antibody to quantify DNA double-strand breaks and the number of y-H2AX foci per cell was determined using Image J. All experiments were carried out in 3 independent experiments. Data are mean ± SD (n > 60 cells per conditions) * <0.05; ** <0.01; *** <0.001. #P<0.05 ### <0.001 compared to untreated shCtrl cells. Representative
Figure 3: Tau inhibition increases mutation frequency measured by the HPRT test. (A) Western-blot analysis of Tau expression in CHO-GFP and CHO-Tau-GFP stable clones using anti -Tau and anti-actin antibodies (B) Schematic representation of the HPRT test in CHO cells. (C) CHO cells were stably transfected with plasmids coding for GFP or GFP-Tau and treated with cisplatin (10 pM, 2 h) or oxaliplatin (20 pM, 2 h). Mutation frequency was measured as the number of colony/number of cells seeded X efficiency of plating. (D) CHO cells were stably transfected with plasmids coding for
GFP or GFP-Tau and treated with X-rays (4 Gy). Mutation frequency was determined as described in B. Data are mean ± SD ***7’0.001..
Figure 4 : Tau increases HR and cNHEJ activities. (A) Schematic representation of DR-GFP reporter. (B) Western-blot analyses of a HeLa DR-GFP stable clone transiently transfected with plasmid encoding the endonuclease I-Scel with or without plasmid encoding Tau protein. (C) Tau increases HR activity. HeLa DR-GFP cells were transfected as described in (B). The percentage of GFP-positive cells was quantitated by flow cytometry (five independent experiments). Results are expressed as fold induction relative to the control. (D) Schematic representation of EJ5GFP reporter. (E) Western-blot analyses of HeLa EJ5-GFP stable clone transiently transfected with plasmid encoding the endonuclease I-Scel with or without plasmid encoding Tau protein. (F) Tau increased cNHEJ activity. HeLa EJ5-GFP cells were transfected as described in (E). The percentage of GFP-positive cells was quantitated by flow cytometry (Five independent experiments). Results are expressed as fold induction relative to the control. Data are mean ± SD **PO.OI; ***7’0.001.
Figure 5. Tau depletion confers sensitivity to doxorubicin and X-rays in mice xenografts. MCF7 cells expressing either scramble (shctrl) or shTau were injected subcutaneously (5 mice per group) and tumor volume was measured for 11 days starting from 100 mm3 and (A) treated or not with doxorubicin (6 mg/kg x 1) or (B) treated or not with X-rays (2 Gy x 2). Average tumor weight ± SD is shown. *7’0.05; **7’0.01; ***7’0.001 compared to untreated cells. ###P<Q.QQ1 compared to treated MCF7 shCtrl xenografts.
Figure 6. Tau increases 53BP1 nuclear translocation after X-ray treatment. (A). MCF7 shCtrl and shTau cells were irradiated with 2 Gy exposure and then further incubated for 15 min. Cells were then labeled with a 53BP1 antibody and the number of 53BP1 foci per cell (n >50) was determined using Image J. All experiments were carried out in 3 independent experiments. Data are mean ± SD 7’0.05; **7’0.01; ***7’0.001. (B) MCF7 shCtrl or shTau stable clones were irradiated with 2 Gy and then further incubated for 15 min. Cells were then labeled with a y-H2AX antibody to quantify DNA double-strand breaks and the number of y-H2AX foci per cell (n >50) was determined using Image J. All experiments were carried out 3 independent experiments. Data are mean ± SD ***7’0.001. (C) The graph shows the quantification of four independent experiments from 53BP1 (red) and y-H2AX (green) staining in MCF7 shCtrl or shTau stable clones irradiated or not with 2 Gy and further incubated for 15 min. Data are mean ± SD *7’0.05; **7’0.01; ***7’0.001. ##7’0.01; ###7’0.001 compared to the corresponding cytoplasmic fraction.
Figure 7. Tau regulates the 53BPl/microtubule interaction. (A) (B) The graph shows the quantification of three independent proximity ligation assays (53BP1-Dynein) experiments as in Proximity Ligation Assay showing 53BP1-Dynein interaction in MCF7shCtrl and shTau cells in control and 2 Gy treated conditions. All data are mean ± SD *7’0.05; **7’0.01; ***7’0.001.
EXAMPLE:
Materials and Methodes
Materials and Plasmids pcDNA3-Tau4R and Tau-GFP have been described elsewhere [14,23,24], pimEJ5GFP was a gift from Jeremy Stark (Addgene plasmid # 44026) [25], pDRGFP (Addgene plasmid # 26475) and pCBAScel (Addgene plasmid # 26477) were a gift from Maria Jasin [26], Short hairpin Tau and RNA Ctrl vectors were purchased from Santacruz. Doxorubicin, cisplatin, oxaliplatin, 6-thioguanine and hypoxanthine were purchased from Sigma-Aldrich and bleomycin from Calbiochem. Cells were exposed to ionizing radiation (IR) using an X-ray machine (Clinac23X).
Cell Culture and Transfection
HeLa cells, MCF7, and MDA-MB-231 were cultured in Dulbecco’s Modified Eagle’s Medium with 10% fetal bovine serum, 2 mM L-glutamine and 50 U/ml penicillin/ streptomycin (Gibco) at 37°C in 5% CO2 humidified air. CHO-K1 cells were cultured in RPMI1640 (Gibco) with 10% fetal bovine serum, 2 mM L-glutamine and 50 U/ml penicillin/streptomycin (Gibco). MCF7 shctrl and shTau cells have been described previously [14], Transient and stable transfection experiments were performed using the lipofectamine LTX reagent according to the manufacturer guideline (Invitrogen). To isolate stably transfected HeLa clones, cells were transfected with the pDRGFP or pimEJ5GFP plasmids and selected with puromycin (2 Dg/ml). Clones were isolated and tested for GFP expression after pCBAScel transfection. Of 6 clones tested for GFP, one was chosen for further studies. For CHO-K1 GFP or GFP-Tau, cells were transfected with plasmids encoding GFP or GFP-Tau and selected with G418 (200 pg/ml). Stably transfected cells were isolated using flow cytometry and cell sorting (Sony SH800).
Cellular extracts and western-blotting
For the preparation of whole cell extracts, cells were washed twice in ice-cold PBS and scraped in lysis buffer [50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1 mM EDTA, 1 mM EGTA, 1% Triton X-100, protease inhibitor mixture (Roche)]. The lysate was rotated fori h at 4°C and centrifuged for 15 min at 4°C. Nuclear and cytoplasmic extracts were prepared as described previously [24], Briefly, Cells were harvested, washed with PBS and resuspended in buffer A (lOmM Hepes pH=7.8, lOmM KC1, 0,5mM EDTA, ImM DTT, complete protease inhibitors [Roche]). Nuclei were recovered by centrifugation for 10 min at 2000g, resuspended in nuclear lysis buffer (50mM Hepes pH 7.4, 1.5mM MgC12, 420mM NaCl, ImM dithiothreitol [DTT] and complete protease inhibitors [Roche]), and then incubated for 1 h in a rotation wheel at 4°C to extract nuclear proteins.
For western-blotting, proteins were solubilized in an SDS loading buffer and analyzed by SDS-PAGE. Primary antibodies used in Western blotting experiments were directed against Tau Cter [14], 53BP1 (Cell Signaling), H3 (Millipore), Hsp90 (Santa Cruz) and P-actin (Sigma). Secondary antibodies coupled to HRP were from Sigma-Aldrich. Immune complexes were detected using the ECL+ system from Amersham/GE Healthcare and observed with an Image Reader LAS4000 (Fujifilm). Quantification was performed by densitometry using Imaged software.
Cell fractionation into cytosolic and microtubule fractions
Cell fractionation was performed as described previously [27], Briefly, equal amounts of MCF7 shCtrl and shTau cells were recovered in buffer A (80 mM Pipes, pH 6.8, 1 mM MgCh, 2 mM EGTA, 30% glycerol, 0.1% Triton X-100, complete protease inhibitors [Roche]). After ultracentrifugation at 100,000 * g at 21 °C for 18 min, supernatants were collected as cytosolic fractions. The pellets (microtubule fraction) were recovered in RIPA buffer and sonicated. Samples were mixed with LDS buffer and equal volumes were loaded for SDS- PAGE and analyzed by immunoblotting.
Hprt mutant Frequency determination
Hrprt mutant frequency was determined by measuring the clonogenicity of cells, as previously described [28], Briefly, CHO-K1 cells stably transfected with GFP or GFP-Tau were grown in hypoxanthine medium for 5 days to eliminate preexisting Hprt mutants, then 1.5 x 106 cells were plated in 75cm2 flasks. After 24 h of culture, cells were treated or not with cisplatin (10 pM, 2h), oxaliplatin (20 pM, 2h) or X-rays (2 Gy). Cells were maintained for 8 days to allow the expression of the Hrprt mutant phenotype. After this period, 2x105 cells were plated in 100mm petri dishes in a complete medium containing 6pg/mL of 6-thioguanine for the selection of Hprt mutants. In parallel, 200 cells were seeded in 35mm dishes (12 dishes/condition), with non-selective medium. Ten days later, colonies formed in both selective and non-selective media were fixed, stained with 4% Giemsa, and scored. Colony efficiency was expressed as a ratio between the number of colonies and the number of seeded cells. Mutant frequency was expressed as a ratio between the colony efficiency of Hprt mutants and those cells cultured in non-selective medium.
Immunofluorescence
Cells were fixed in 4% paraformaldehyde for 30 min at room temperature. Permeabilization was carried out in 0.2% Triton X-100 in phosphate-buffered saline for 10 min at room temperature. After 1 h saturation in 2% bovine serum albumin, immunostaining was performed using yH2AX (Millipore), 53BP1 (Cell Signaling), y- tubulin (Millipore). These
antibodies were revealed via secondary antibodies coupled to Alexa 488 or 568 (Life Technologies). Nuclear staining was performed by adding 1/2000 DAPI (Img/mL, Life Technologies) in phosphate-buffered saline for 10 min. Slides were then analyzed with a Zeiss LSM710 confocal laser scanning microscope (60x magnification). Images were collected in the Z direction at 0.80 mm intervals and quantified using the ImageJ plug-in as described previously [14],
Proximity ligation assay
Proximity ligation assay was performed with a duolink proximity ligation assay kit (Sigma-Aldrich) according to the manufacturer's guidelines. 53BP1 (Millipore) and dynein (Abeam) were used to visualize 53BP1 -dynein proximity.
DNA repair reporter assays
HeLa cells with stably integrated HR (DR-GFP) or nonhomologous end joining (EJ5GFP) reporters were plated in 6-well plates and then transfected with a plasmid encoding the LScel endonuclease gene with or without Tau. The cells were left to grow for 48 h and then trypsinized and washed once with PBS. GFP fluorescence was analyzed using an LSR FORTESSA X20 cytometer (Becton Dickinson).
Xenograft Studies
The animals were maintained in compliance with European standards for the care and use of laboratory animals and experimental protocols approved by the local Animal Ethical Committee (agreement APAFIS# 27259-2020091810585608, Lille, France). 10 million MCF7, MCF7 shCtrl or MCF7 shTau cells were suspended in 100 pL of FBS/Matrigel v/v (Invitrogen), and then injected in the fat pad of 6-week-old female NOD/SCID mice (Jackson Laboratory). Tumors were measured with calipers, calculated by the formula volume= L x I2 x A. When tumors reached - 100 mm3, mice were assigned randomly to either control, doxorubicin (6 mg/kg once via intravenous injection) or X-rays (N=10 tumors). Mice were then treated with PBS or doxorubicin (6 mg/kg). For X-rays, mice were anesthetized (ketamine, 100 mg/kg and xylazine, 20 mg/kg) and the tumor site was then exposed to radiation (2 Gy x 2) using an XRAD 320. Tumors are measured with calipers and volume was calculated as described previously.
Statistical Analysis
Data are mean ± SD. Statistical tests were carried out using GraphPad Prism software (GraphPad Inc.). Statistical significance between groups was analyzed with Wilcoxon- Mann- Whitney test and Mann-Whitney tests. A p-value less than 0.05 was considered significant.
Results
Tau aids clearance of double-strand breaks.
To assess a potential role of Tau in protecting the DNA of cancer cells, we used MCF7 and MDA-MD-231 cells stably overexpressing either small hairpin RNA control (shCtrl) or targeting Tau (shTau) in which we observed 70% decrease in Tau expression [14], These control and Tau knockdown cells were treated for 2h with bleomycin (30 pg/ml), a drug known to induce double-strand breaks. We then quantified DSBs using a phosphorylated form of H2AX as an indicator of DNA damage[29]. Where we knocked down Tau, y-H2AX fluorescence intensity increased 4-fold compared to just 2-fold in both MCF7 and MDA-MD- 231 control cells (Figures 1A and IB).
Next we tested the effect of knocking down Tau on DSB clearance kinetics after a pulse of X irradiation. MCF7 and MDA-MD-231 clones were irradiated with 2 Gy and the number of y-H2AX foci was then quantified at 5 min, 2, 4 and 6h post-irradiation. A 2-fold increase in DSB foci in both MCF7-shCtrl and shTau cells clones was observed. However, while the level of y-H2AX foci decreased after 2h and had nearly returned to its initial level after 6h in MCF7- shCtrl cells, it remained unchanged 6h post-irradiation in MCF7-shTau cells (Figure 2A). We obtained similar results using another shRNA directed against Tau ruling out possible of target effects (data not shown). The same trends were observed in MDA-MD-231 shCtrl and shTau cells (Figure 2B).
Altogether, these observations suggest a role for Tau in DSB clearance/repair.
Tau decreases the mutation rate induced by DNA damaging agents
The above results suggested that Tau could be a factor that mediates resistance to DNA damaging agents. We therefore performed a classic Hprt mutation test to assess the rates of spontaneous or cisplatin-, oxaliplatin- and X-ray-induced mutations in the absence or presence of Tau. CHO KI cells, which possess only one functional Hprt allele, were stably transfected with plasmids encoding GFP or GFP-Tau fusion protein, then treated with cisplatin, oxaliplatin or X-rays (Figure 3 A). The addition of 6-thioguanine allows the selection of Hprt mutants, which reflect the mutation frequencies (Figure 3B). Figure 3C and 3D demonstrate that the frequency of spontaneous 6- thioguanine resistant mutants was not significantly different in CHO KI cells expressing Tau compared to GFP control cells. To study if Tau reduced the frequency of DNA damaging agents-induced mutations at the Hprt locus, GFP or GFP-Tau CHO KI cells were treated with cisplatin (10 pM, 2 h), oxaliplatin (20 pM, 2 h) or X-rays (4 Gy). In the three cases, the frequencies of 6- thioguanine induced by these compounds were significantly lower in cells expressing Tau (3.3-, 3.1- and 2.8-fold for cisplatin, oxaliplatin and X-rays respectively).
Thus, these results confirmed a significant role for Tau in the repair of anticancer agent induced-DNA lesions.
Tau increases HR and cNHEJ activities.
The repair of DSB breaks in mammalian cells occurs mainly by the cNHEJ or the HR pathway. To ascertain if Tau acts on a specific DSB pathway, we employed reporter genes of either HR or cNHEJ. Firstly we employed the HR reporter gene (DR-GFP), which contains a non-functional GFP due to the replacement of 11 bp of the GFP sequence by the rare cutting I- Scel endonuclease recognition site. The I-Scel endonuclease generates a DSB. The HR repair restores the proper GFP coding sequence, resulting in GFP expression which is further quantified by flow cytometry (Figure 4A) [24], We established a HeLa DR-GFP stable clone and then transiently transfected a plasmid encoding the I-Scel endonuclease in the absence (Ctrl) or presence of plasmid encoding Tau (Tau). The endonuclease expression level was verified and shown to be similar between HeLa-DR-GFP-Ctrl and HeLa-DR-GFP-Tau cells (Figure 4B). Noticeably, Tau overexpression resulted in a 1.5-fold increase in the number of cells expressing GFP in HeLa-DR-GFP-Tau (Figure 4C). These results indicated that Tau promoted a more efficient HR repair.
Secondly, we evaluated the cNHEJ DNA repair process using the EJ5-GFP reporter [23], In this plasmid, the promoter is separated from the GFP by a puromycin gene containing two I-Scel sites at the 5’ and 3’ positions. Following I-Scel cleavage, the puromycin gene is excised, leading to the restoration of the GFP signal by NHEJ repair (Figure 4D). A selected HeLa EJ5-GFP clone was therefore transfected with a plasmid encoding the I-Scel in the absence or presence of plasmid encoding Tau. As above, the level of I-Scel expression was similar between the two conditions (Figure 4E). As for HR also, we observed a 1.6-fold increase in cNHEJ repair activity when Tau was expressed (Figure 4F).
Taken together, these data demonstrated that Tau increased both HR and cNHEJ repair efficiency. shTau tumors are more sensitive to DNA damage.
Finally, we sought to test the in vivo relevance of our cellular findings. For this, MCF7- shCtrl and shTau cells were injected subcutaneously into the mammary fat pads of immunodeficient SCID mice. The tumor volumes were measured every three days until they reached 100 mm3 then treated with X-rays or the widely used chemotherapy drug, doxorubicin. As shown in Figure 5A, there were no differences in the tumor growth between shCtrl and shTau xenografts during these 11 days in the absence of doxorubicin. When treated with doxorubicin, shCtrl xenografts remained at approximately the starting volume, neither growing
nor shrinking. For shTau xenografts however, doxorubicin treatment led to decrease in tumor volume, of 12.7% and 28.1% on days 5 and 11 respectively (Figure 5 A). Similar results were observed after x ray treatment with X rays reducing tumor volume more in the shTau xenografts with a 29.2% and 30.2% decrease on days 5 and 11 respectively (Figure 5B).
Together, these data strongly support that Tau inhibition potentiates the therapeutic effect of doxorubicin and X-rays by inhibiting DNA repair.
Tau regulates 53BP1 nuclear localization.
While we observed that Tau increased both cNHEJ and HR, it is known that cNHEJ accounts formost ofDSB repair in mammalian cells [7,8], In addition, the recruitment of 53BP1 at DSBs is known to be critical for this mechanism [6],
We next examined the formation of 53BP1 foci in MCF7 shCtrl and shTau cells by immufluorescence. As shown in Figure 6A and 6C, we observed a 4.5-fold increase in the number of 53BP1 foci in shCtrl cells 15 min after 2 Gy irradiation. However, knocking-down Tau expression led to a decrease in 53BP1 foci number (2.1 -fold increase 15 min after 2 Gy irradiation). On the other hand Tau knockdown had no effect on the number of y-H2AX foci (Figure 6B). Similar results were found 30 min after 2 Gy irradiation.
Lower abundance of nuclear 53BP1 foci was not explained by an expression change. We therefore hypothesized that the observed differences in shCtrl and shTau conditions could be due to a perturbed nuclear localization. Indeed, it has been demonstrated that efficient DSB repair relies on nuclear translocation of DNA repair proteins such as 53BP1 [9], To test this, intracellular 53BP1 distribution was then tested using subcellular fractionation (Figure 6C). For MCF7 shCtrl cells, 53BP1 protein was found primarily in the nuclear fraction in control (66%) and irradiated conditions (75%). Although the distribution was similar in the MCF7 shTau cells in control conditions (60% in the nuclear fraction), we observed a strong decrease in the nuclear fraction after X-rays treatment (30%).
These data therefore suggest that Tau regulates the cyto-nuclear shuttling of 53BP1
Tau silencing alters 53BP1 trafficking on microtubules.
It has been shown that nuclear translocation of 53BP1 and other DNA repair proteins, relies in part on their interaction with the retrograde motor protein dynein [9], Since Tau is mostly known as a microtubule-binding protein that regulate dynein, we next wondered if its absence could impede the interaction of 53BP1 with microtubules. We first compared the microtubule-association of 53BP1. For this, 53BP1 distribution between the cytosolic and microtubule fraction was assessed in control or X-ray treated MCF7-shCtrl or -shTau cells. As shown in Figure 7A, 53BP1 is present in both cytoplasmic and microtubule fractions.
Noticeably, we observed a 2-fold difference in 53BP1 -microtubule complex enrichment in nontreated MCF7-shTau cells when compared to non-treated MCF7-shCtrl. Moreover, we found that 53BPl/microtubule association was further increased in MCF7shCtrl cells after 2 Gy irradiation (1.9-fold increase) while decreasing in MCF7 shTau cells (2.1-fold decrease).
Since the higher association could indicated either an increase or a stronger association of 53BP1 with microtubules, we next examined whether there were also differences in 53BPl/dynein interaction. For this, a proximity ligation assay was performed to specifically detect 53BPl/dynein interaction in MCF7-shCtrl and shTau cells in control or 2 Gy treated conditions. As shown in Figures 7B, proximity was increased in shCtrl cells after irradiation (2.3-fold) while the signal was lowered in MCF7-shTau cells (1.2-fold increase).
Discussion
In this work, we investigated the role of Tau in repairing DNA double-strand breaks in cancer cells. We report a new function of Tau in DSB repair that it carries out by regulating DNA repair protein trafficking. Notably, we identified Tau as a major actor in regulating cNHEJ through regulating 53BP1 trafficking on microtubules
This finding has precedents in several studies suggesting a relationship between Tau and DNA damage [15,30], In particular, an increase in y-H2AX foci was observed under stress conditions, in primary neuronal cell culture in the absence of Tau or in the cortex and hippocampus of knock-out Tau mice [31,32], Although it has been suggested that Tau binds and protects DNA from damage, our observations do not support this hypothesis. First, the same level of H2AX phosphorylation was observed in shCtrl and shTau cells after irradiation, which seems to be incompatible with a direct DNA protective role for Tau. Second, we observed a delay in the resolution of □-H2AX foci in shTau cells that further suggested a role for Tau in DNA repair as opposed to DNA binding per se.
The observed kinetics of y-H2AX resolution suggested that Tau mostly assists DNA repair through cNHEJ pathway. To obtain more detailed insights into the Tau function in cNHEJ, we examined the formation of 53BP1 foci in shCtrl and shTau cells. A lower 53BP1 recruitment to DNA damage sites is known to be sufficient to hinder DSB repair by cNHEJ [33], However, while most previous studies report a decrease in 53BP1 expression upon degradation, we do not observe variations in protein level but rather an alteration in protein localization. Dysregulation of nucleocytoplasmic transport has previously been implicated in neuronal Tau pathology. Protein aggregates have been shown to interact with different components of the nuclear pore complex and alter nucleocytoplasmic transport [34,35], Using Frontotemporal Lobar Degeneration Tau mutations (FTDP-MAPT), others demonstrated that
changes in microtubule dynamics caused nuclear envelope distortions and marked disruption of nucleocytoplasmic transport [36], These observations suggest that Tau loss-of-function impedes microtubule nuclear translocation.
Our results therefore suggest that 53BP1 nucleocytoplasmic shuttling is highly dependent on Tau. 53BP1 is present both in the cytoplasm and nucleus in shCtrl cells and the nuclear translocation of 53BP1 tends to increase after DSB induction. Furthermore we observed increased interaction of 53BP1 with both dynein and microtubules in the presence of Tau when treated with DNA damaging agents. Interestingly, it has been observed an enhanced nuclear targeting of the human adenovirus type 2 (Ad2) after overexpression of MAP4 and relying on directed transport along microtubules by dynein [37], These observations further suggest that Tau might exert a similar role on dynein-directed protein translocation. Indeed, Tau depletion led to strong alterations in dynein-directed protein translocation. In shTau control cells, we observed an increase in the 53BP1 microtubule fraction independently of nuclear translocation. Although the current explanation is not known, this might reflect a lower displacement rate of dynein complex along microtubules. Tau is known to increase the frequency of long dynein- directed runs [38], Altogether, these observations are in agreement with previous results showing that altering microtubule dynamics with vincristine and paclitaxel increased 53BP1 cytoplasmic retention and accentuated the effect of DNA damaging agents [9],
It is important to note that an alteration in 53BP1 protein localization is observed after DSB induction, suggesting that microtubule dynamics are regulated by DSBs themselves. It has been demonstrated that upon double-strand breakage, cytoplasmic microtubules invade the nucleus [39], This mechanism is thought to increase the mobility of damaged DNA inside the nucleus necessary for DNA repair [10,40,41], Interestingly, it has been shown recently in neurons that DSBs increased the interaction between Tau and tubulin around the nuclear membrane, and further suggesting the enhancement of tubulin polymerization [32], Based on our observations, it is tempting to speculate that Tau, responds to DSBs, by enhancing microtubule polymerization and/or stability necessary for DNA mobility and nuclear translocation of DNA repair proteins.
Transport of DNA repair proteins along microtubules is not restricted to 53BP1 but also to other proteins involved in different DNA repair pathways. p53, DNA-PK complex proteins (Ku70, Ku80, DNA-PKcs), BRCA1/2, MRN complex proteins (Mrel 1, Rad50, Nbsl), or RPA and Rad51 are also subjected to microtubule trafficking [9,42-44], In this regard, we found that the efficiency of the HR pathway in reporter gene assay is increased in the presence of Tau. This observation further suggested that Tau did not impact on DSB repair pathways choice. In
this regard, we also show that Tau expressing cells are less sensitive to cisplatin and oxaliplatin treatments. Platinum drugs acts mainly by forming intrastrand diadducts, primarily repaired via the nucleotide excision repair system [45], Interestingly, cytosolic sequestration of DNA repair proteins has been linked to neuronal death in different Tauopathies such as Alzheimer’s disease, Pick’s disease, corticobasal neurodegeneration or progressive supranuclear palsy [46,47],
Conclusions
In this work, we provide evidence for a new role of Tau in DNA repair, promoting resistance to commonly used anti-cancer treatments. These findings further suggest that Tau expression may be of interest as a molecular marker for response to DNA damaging agents and as a beneficial therapeutic target in tumors.
REFERENCES:
Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.
1. Trenner, A.; Sartori, A. A. Harnessing DNA double-strand break repair for cancer treatment. Front Oncol 2019, 9, 1388.
2. Khanna, K.K.; Jackson, S.P. DNA double-strand breaks: Signaling, repair and the cancer connection. Nat Genet 2001, 7, 247-254.
3. Her, J.; Bunting, S.F. How cells ensure correct repair of DNA double-strand breaks. J Biol Ghent 2018, 293, 10502-10511.
4. Ceccaldi, R.; Rondinelli, B.; D'Andrea, A.D. Repair pathway choices and consequences at the double-strand break. Trends Cell Biol 2016, 26, 52-64.
5. Clouaire, T.; Legube, G. A snapshot on the cis chromatin response to DNA doublestrand breaks. Trends Genet 2019, 35, 330-345.
6. Chapman, J.R.; Sossick, A.J.; Boulton, S.J.; Jackson, S.P. Brcal -associated exclusion of 53bpl from DNA damage sites underlies temporal control of DNA repair. J Cell Sci 2012, 125, 3529-3534.
7. Mao, Z.; Bozzella, M.; Seluanov, A.; Gorbunova, V. Comparison of nonhomologous end joining and homologous recombination in human cells. DNA Repair (Amst) 2008, 7, 1765- 1771.
8. Beucher, A.; Birraux, J.; Tchouandong, L.; Barton, O.; Shibata, A.; Conrad, S.; Goodarzi, A.A.; Krempler, A.; Jeggo, P.A.; Lobrich, M. Atm and artemis promote homologous
recombination of radiation-induced DNA double-strand breaks in g2. EMBO J2009, 28, 3413- 3427.
9. Poruchynsky, M.S.; Komlodi-Pasztor, E.; Trostel, S.; Wilkerson, J.; Regairaz, M.; Pommier, Y.; Zhang, X.; Kumar Maity, T.; Robey, R.; Burotto, M., etal. Microtubule-targeting agents augment the toxicity of DNA-damaging agents by disrupting intracellular trafficking of DNA repair proteins. Proc Natl Acad Set U S A 2015, 112, 1571-1576.
10. Lottersberger, F.; Karssemeijer, R.A.; Dimitrova, N.; de Lange, T. 53bpl and the line complex promote microtubule-dependent dsb mobility and DNA repair. Cell 2015, 163, 880- 893.
11. Weingarten, M.D.; Lockwood, A.H.; Hwo, S.Y.; Kirschner, M.W. A protein factor essential for microtubule assembly. Proc Natl Acad Sci U SA 1975, 72, 1858-1862.
12. Sotiropoulos, I.; Galas, M.C.; Silva, J.M.; Skoulakis, E.; Wegmann, S.; Maina, M.B.; Blum, D.; Sayas, C.L.; Mandelkow, E.M.; Mandelkow, E., et al. Atypical, non-standard functions of the microtubule associated tau protein. Acta Neuropathol Commun 2017, 5, 91.
13. Frost, B.; Hemberg, M.; Lewis, J.; Feany, M.B. Tau promotes neurodegeneration through global chromatin relaxation. Nat Neurosci 2014, 17, 357-366.
14. Rico, T.; Gilles, M.; Chauderlier, A.; Comptdaer, T.; Magnez, R.; Chwastyniak, M.; Drobecq, H.; Pinet, F.; Thuru, X.; Buee, L., et al. Tau stabilizes chromatin compaction. Front Cell Dev Biol 2021, 9, 740550.
15. Colnaghi, L.; Rondelli, D.; Muzi-Falconi, M.; Sertic, S. Tau and DNA damage in neurodegeneration. Brain Sci 2020, 10.
16. Violet, M.; Delattre, L.; Tardivel, M.; Sultan, A.; Chauderlier, A.; Caillierez, R.; Talahari, S.; Nesslany, F.; Lefebvre, B.; Bonnefoy, E., et al. A major role for tau in neuronal DNA and rna protection in vivo under physiological and hyperthermic conditions. Front Cell Neurosci 2014, 8, 84.
17. Violet, M.; Chauderlier, A.; Delattre, L.; Tardivel, M.; Chouala, M.S.; Sultan, A.; Marciniak, E.; Humez, S.; Binder, L.; Kayed, R., et al. Prefibrillar tau oligomers alter the nucleic acid protective function of tau in hippocampal neurons in vivo. Neurobiol Dis 2015, 82, 540-551.
18. Wei, Y.; Qu, M.H.; Wang, X.S.; Chen, L.; Wang, D.L.; Liu, Y.; Hua, Q.; He, R.Q. Binding to the minor groove of the double-strand, tau protein prevents DNA from damage by peroxidation. PLoS One 2008, 3, e2600.
19. Qi, H.; Cantrelle, F.X.; Benhelli-Mokrani, H.; Smet-Nocca, C.; Buee, L.; Lippens, G.; Bonnefoy, E.; Galas, M.C.; Landrieu, I. Nuclear magnetic resonance spectroscopy
characterization of interaction of tau with DNA and its regulation by phosphorylation. Biochemistry 2015, 54, 1525-1533.
20. Hedna, R.; Kovacic, H.; Pagano, A.; Peyrot, V.; Robin, M.; Devred, F.; Breuzard, G. Tau protein as therapeutic target for cancer? Focus on glioblastoma. Cancers (Basel) 2022, 14.
21. Rossi, G.; Redaelli, V.; Perego, P.; Ferrari, R.; Giaccone, G.; Tagliavini, F. Tau mutations as a novel risk factor for cancer-response. Cancer Res 2018, 78, 6525.
22. Papin, S.; Paganetti, P. Emerging evidences for an implication of the neurodegeneration- associated protein tau in cancer. Brain Sci 2020, 10.
23. Delobel, P.; Flament, S.; Hamdane, M.; Jakes, R.; Rousseau, A.; Delacourte, A.; Vilain, J.P.; Goedert, M.; Buee, L. Functional characterization of ftdp-17 tau gene mutations through their effects on xenopus oocyte maturation. J Biol Chem 2002, 277, 9199-9205.
24. Chauderlier, A.; Gilles, M.; Spolcova, A.; Caillierez, R.; Chwastyniak, M.; Kress, M.; Drobecq, H.; Bonnefoy, E.; Pinet, F.; Weil, D., et al. Tau/ddx6 interaction increases microrna activity. Biochim Biophys Acta Gene Regul Meeh 2018, 1861, 762-772.
25. Bennardo, N.; Cheng, A.; Huang, N.; Stark, J.M. Alternative-nhej is a mechanistically distinct pathway of mammalian chromosome break repair. PLoS Genet 2008, 4, el 000110.
26. Pierce, A.J.; Johnson, R.D.; Thompson, L.H.; Jasin, M. Xrcc3 promotes homology- directed repair of DNA damage in mammalian cells. Genes Dev 1999, 13, 2633-2638.
27. Derisbourg, M.; Leghay, C.; Chiappetta, G.; Fernandez-Gomez, F.J.; Laurent, C.; Demeyer, D.; Carrier, S.; Buee-Scherrer, V.; Blum, D.; Vinh, J., etal. Role of the tau n-terminal region in microtubule stabilization revealed by new endogenous truncated forms. Sci Rep 2015, 5, 9659.
28. Silva, M.J.; Costa, P.; Dias, A.; Valente, M.; Louro, H.; Boavida, M.G. Comparative analysis of the mutagenic activity of oxaliplatin and cisplatin in the hprt gene of cho cells. Environ Mol Mutagen 2005, 46, 104- 115.
29. Atkinson, J.; Bezak, E.; Kempson, I. Imaging DNA double-strand breaks - are we there yet? Nat Rev Mol Cell Biol 2022, 23, 579-580.
30. Galas, M.C.; Bonnefoy, E.; Buee, L.; Lefebvre, B. Emerging connections between tau and nucleic acids. Adv Exp Med Biol 2019, 1184, 135-143.
31. Sultan, A.; Nesslany, F.; Violet, M.; Begard, S.; Loyens, A.; Talahari, S.; Mansuroglu, Z.; Marzin, D.; Sergeant, N.; Humez, S., et al. Nuclear tau, a key player in neuronal DNA protection. J Biol Chem 2011, 286, 4566-4575.
32. Asada-Utsugi, M.; Uemura, K.; Ayaki, T.; M, T.U.; Minamiyama, S.; Hikiami, R.; Morimura, T.; Shodai, A.; Ueki, T.; Takahashi, R., et al. Failure of DNA double-strand break repair by tau mediates alzheimer's disease pathology in vitro. Commun Biol 2022, 5, 358.
33. Gonzalez-Suarez, I.; Redwood, A.B.; Grotsky, D.A.; Neumann, M.A.; Cheng, E.H.; Stewart, C.L.; Dusso, A.; Gonzalo, S. A new pathway that regulates 53bpl stability implicates cathepsin 1 and vitamin d in DNA repair. EMBO J 2011, 30, 3383-3396.
34. Lester, E.; Parker, R. The tau of nuclear-cytoplasmic transport. Neuron 2018, 99, 869- 871.
35. Diez, L.; Wegmann, S. Nuclear transport deficits in tau-related neurodegenerative diseases. Front Neurol 2020, 11, 1056.
36. Paonessa, F.; Evans, L.D.; Solanki, R.; Larrieu, D.; Wray, S.; Hardy, J.; Jackson, S.P.; Livesey, F.J. Microtubules deform the nuclear membrane and disrupt nucleocytoplasmic transport in tau-mediated frontotemporal dementia. Cell Rep 2019, 26, 582-593 e585.
37. Suomalainen, M.; Nakano, M.Y.; Keller, S.; Boucke, K.; Stidwill, R.P.; Greber, U.F. Microtubule-dependent plus- and minus end-directed motilities are competing processes for nuclear targeting of adenovirus. J Cell Biol 1999, 144, 657-672.
38. Chaudhary, A.R.; Berger, F.; Berger, C.L.; Hendricks, A.G. Tau directs intracellular trafficking by regulating the forces exerted by kinesin and dynein teams. Traffic 2018, 19, 111- 121.
39. Oshidari, R.; Strecker, J.; Chung, D.K.C.; Abraham, K.J.; Chan, J.N.Y.; Damaren, C.J.; Mekhail, K. Nuclear microtubule filaments mediate non-linear directional motion of chromatin and promote DNA repair. Nat Commun 2018, 9, 2567.
40. Mekhail, K. Defining the damaged DNA mobility paradox as revealed by the study of telomeres, dsbs, microtubules and motors. Front Genet 2018, 9, 95.
41. Mekhail, K. Corrigendum: Defining the damaged DNA mobility paradox as revealed by the study of telomeres, dsbs, microtubules and motors. Front Genet 2018, 9, 157.
42. Giannakakou, P.; Sackett, D.L.; Ward, Y.; Webster, K.R.; Blagosklonny, M.V.; Fojo, T. P53 is associated with cellular microtubules and is transported to the nucleus by dynein. Nat Cell Biol 2000, 2, 709-717.
43. Giannakakou, P.; Nakano, M.; Nicolaou, K.C.; O'Brate, A.; Yu, J.; Blagosklonny, M.V.; Greber, U.F.; Fojo, T. Enhanced microtubule-dependent trafficking and p53 nuclear accumulation by suppression of microtubule dynamics. Proc Natl Acad Sci U S A 2002, 99, 10855-10860.
44. Li, Q.; Martinez, J.D. P53 is transported into the nucleus via an hsfl -dependent nuclear localization mechanism. Mol Carcinog 2011, 50, 143-152.
45. Basu, A.; Krishnamurthy, S. Cellular responses to cisplatin-induced DNA damage. J Nucleic Acids 2010, 2010. 46. Kurihara, M.; Mano, T.; Saito, Y.; Murayama, S.; Toda, T.; Iwata, A. Colocalization of brcal with tau aggregates in human tauopathies. Brain Sci 2019, 10.
47. Nakamura, M.; Kaneko, S.; Dickson, D.W.; Kusaka, H. Aberrant accumulation of brcal in alzheimer disease and other tauopathies. J Neuropathol Exp Neurol 2020, 79, 22-33.
Claims
1. A method for decreasing therapeutic resistance to chemotherapy agent and/or radiotherapy agent in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a Tau inhibitor.
2. A method for preventing and/or treating cancer with acquired resistance to treatment with chemotherapy agent and/or radiotherapy agent in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a Tau inhibitor.
3. The method according to claims 1 or 2, wherein the Tau inhibitor is
A) Inhibitor of Tau activity selected from the list consisting Anti-Tau antibody and anti-Tau aptamers, tau peptide (vaccines)
B) PROTAC (“Proteolysis Targeting Chimera”) which simultaneously bind a tau protein and an E3-ubiquitin ligase.
C) Inhibitor of Tau gene expression selected from the list consisting of antisense, oligonucleotide, nuclease, siRNA, shRNA or ribozyme nucleic acid sequence.
4. . The method according to claim 3 wherein the tau Inhibitor is an anti-Tau antibody.
5. The method according to claim 2 wherein the cancer is selected from the list consisting in breast cancer, gastric, cancer prostate cancer ovarian cancer, glioma, Neuroblastoma, Kidney clear cell carcinoma, Lung cancer, Pheochromocytoma/Paraganglioma and Neuroendocrine tumors (NETs).
6. The method according to claims 1 to 6 wherein the chemotherapeutic agent is cisplatin, doxorubicin, carboplatin or oxaliplatin.
7. The method according to claims 1 to 6 wherein the radiotherapeutic agent is X-rays.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP22306999.8 | 2022-12-22 | ||
EP22306999 | 2022-12-22 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024133723A1 true WO2024133723A1 (en) | 2024-06-27 |
Family
ID=85017760
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2023/087320 WO2024133723A1 (en) | 2022-12-22 | 2023-12-21 | Methods for decreasing therapeutic acquired resistance to chemotherapy and/or radiotherapy |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024133723A1 (en) |
Citations (35)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US541A (en) | 1837-12-26 | Daniel desmond | ||
US5874A (en) | 1848-10-24 | Apparatus eob baking- water | ||
US4816397A (en) | 1983-03-25 | 1989-03-28 | Celltech, Limited | Multichain polypeptides or proteins and processes for their production |
US4816567A (en) | 1983-04-08 | 1989-03-28 | Genentech, Inc. | Recombinant immunoglobin preparations |
EP0368684A1 (en) | 1988-11-11 | 1990-05-16 | Medical Research Council | Cloning immunoglobulin variable domain sequences. |
US4946778A (en) | 1987-09-21 | 1990-08-07 | Genex Corporation | Single polypeptide chain binding molecules |
WO1993011161A1 (en) | 1991-11-25 | 1993-06-10 | Enzon, Inc. | Multivalent antigen-binding proteins |
US5225539A (en) | 1986-03-27 | 1993-07-06 | Medical Research Council | Recombinant altered antibodies and methods of making altered antibodies |
US5800988A (en) | 1992-08-21 | 1998-09-01 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
US5981732A (en) | 1998-12-04 | 1999-11-09 | Isis Pharmaceuticals Inc. | Antisense modulation of G-alpha-13 expression |
US6046321A (en) | 1999-04-09 | 2000-04-04 | Isis Pharmaceuticals Inc. | Antisense modulation of G-alpha-i1 expression |
US6075181A (en) | 1990-01-12 | 2000-06-13 | Abgenix, Inc. | Human antibodies derived from immunized xenomice |
US6107091A (en) | 1998-12-03 | 2000-08-22 | Isis Pharmaceuticals Inc. | Antisense inhibition of G-alpha-16 expression |
US6150584A (en) | 1990-01-12 | 2000-11-21 | Abgenix, Inc. | Human antibodies derived from immunized xenomice |
US6365354B1 (en) | 2000-07-31 | 2002-04-02 | Isis Pharmaceuticals, Inc. | Antisense modulation of lysophospholipase I expression |
US6410323B1 (en) | 1999-08-31 | 2002-06-25 | Isis Pharmaceuticals, Inc. | Antisense modulation of human Rho family gene expression |
US6566135B1 (en) | 2000-10-04 | 2003-05-20 | Isis Pharmaceuticals, Inc. | Antisense modulation of caspase 6 expression |
US6566131B1 (en) | 2000-10-04 | 2003-05-20 | Isis Pharmaceuticals, Inc. | Antisense modulation of Smad6 expression |
US6765087B1 (en) | 1992-08-21 | 2004-07-20 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
US6838254B1 (en) | 1993-04-29 | 2005-01-04 | Conopco, Inc. | Production of antibodies or (functionalized) fragments thereof derived from heavy chain immunoglobulins of camelidae |
US6982321B2 (en) | 1986-03-27 | 2006-01-03 | Medical Research Council | Altered antibodies |
WO2006003388A2 (en) | 2004-06-30 | 2006-01-12 | Domantis Limited | Compositions and methods for treating inflammatory disorders |
WO2006030220A1 (en) | 2004-09-17 | 2006-03-23 | Domantis Limited | Compositions monovalent for cd40l binding and methods of use |
US7087409B2 (en) | 1997-12-05 | 2006-08-08 | The Scripps Research Institute | Humanization of murine antibody |
WO2012049570A1 (en) | 2010-10-11 | 2012-04-19 | Panima Pharmaceuticals Ag | Human anti-tau antibodies |
US20140068797A1 (en) | 2012-05-25 | 2014-03-06 | University Of Vienna | Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription |
US8697359B1 (en) | 2012-12-12 | 2014-04-15 | The Broad Institute, Inc. | CRISPR-Cas systems and methods for altering expression of gene products |
WO2014096321A1 (en) | 2012-12-21 | 2014-06-26 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Antibodies specific to tau phosphorylated at serine 422 and uses for the treatment and diagnosis of tauopathies |
WO2015004163A1 (en) | 2013-07-08 | 2015-01-15 | Adx Neurosciences | Anti-tau monoclonal antibodies |
WO2015200806A2 (en) | 2014-06-27 | 2015-12-30 | C2N Diagnostics Llc | Humanized anti-tau antibodies |
WO2016112078A2 (en) | 2015-01-08 | 2016-07-14 | Janssen Biotech, Inc. | Anti-phf-tau antibodies and their uses |
WO2018152359A1 (en) | 2017-02-17 | 2018-08-23 | Denali Therapeutics Inc. | Anti-tau antibodies and methods of use thereof |
WO2020120644A1 (en) | 2018-12-13 | 2020-06-18 | INSERM (Institut National de la Santé et de la Recherche Médicale) | New anti tau svqivykpv epitope single domain antibody |
WO2020193520A1 (en) | 2019-03-25 | 2020-10-01 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Treatment of taupathy disorders by targeting new tau species |
WO2021010712A1 (en) | 2019-07-15 | 2021-01-21 | 주식회사 아델 | Anti-tau antibody and use of same |
-
2023
- 2023-12-21 WO PCT/EP2023/087320 patent/WO2024133723A1/en unknown
Patent Citations (36)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5874A (en) | 1848-10-24 | Apparatus eob baking- water | ||
US541A (en) | 1837-12-26 | Daniel desmond | ||
US4816397A (en) | 1983-03-25 | 1989-03-28 | Celltech, Limited | Multichain polypeptides or proteins and processes for their production |
US4816567A (en) | 1983-04-08 | 1989-03-28 | Genentech, Inc. | Recombinant immunoglobin preparations |
US5225539A (en) | 1986-03-27 | 1993-07-06 | Medical Research Council | Recombinant altered antibodies and methods of making altered antibodies |
US6982321B2 (en) | 1986-03-27 | 2006-01-03 | Medical Research Council | Altered antibodies |
US4946778A (en) | 1987-09-21 | 1990-08-07 | Genex Corporation | Single polypeptide chain binding molecules |
EP0368684A1 (en) | 1988-11-11 | 1990-05-16 | Medical Research Council | Cloning immunoglobulin variable domain sequences. |
US6075181A (en) | 1990-01-12 | 2000-06-13 | Abgenix, Inc. | Human antibodies derived from immunized xenomice |
US6150584A (en) | 1990-01-12 | 2000-11-21 | Abgenix, Inc. | Human antibodies derived from immunized xenomice |
WO1993011161A1 (en) | 1991-11-25 | 1993-06-10 | Enzon, Inc. | Multivalent antigen-binding proteins |
US6015695A (en) | 1992-08-21 | 2000-01-18 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
US5800988A (en) | 1992-08-21 | 1998-09-01 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
US6765087B1 (en) | 1992-08-21 | 2004-07-20 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
US6838254B1 (en) | 1993-04-29 | 2005-01-04 | Conopco, Inc. | Production of antibodies or (functionalized) fragments thereof derived from heavy chain immunoglobulins of camelidae |
US7087409B2 (en) | 1997-12-05 | 2006-08-08 | The Scripps Research Institute | Humanization of murine antibody |
US6107091A (en) | 1998-12-03 | 2000-08-22 | Isis Pharmaceuticals Inc. | Antisense inhibition of G-alpha-16 expression |
US5981732A (en) | 1998-12-04 | 1999-11-09 | Isis Pharmaceuticals Inc. | Antisense modulation of G-alpha-13 expression |
US6046321A (en) | 1999-04-09 | 2000-04-04 | Isis Pharmaceuticals Inc. | Antisense modulation of G-alpha-i1 expression |
US6410323B1 (en) | 1999-08-31 | 2002-06-25 | Isis Pharmaceuticals, Inc. | Antisense modulation of human Rho family gene expression |
US6365354B1 (en) | 2000-07-31 | 2002-04-02 | Isis Pharmaceuticals, Inc. | Antisense modulation of lysophospholipase I expression |
US6566131B1 (en) | 2000-10-04 | 2003-05-20 | Isis Pharmaceuticals, Inc. | Antisense modulation of Smad6 expression |
US6566135B1 (en) | 2000-10-04 | 2003-05-20 | Isis Pharmaceuticals, Inc. | Antisense modulation of caspase 6 expression |
WO2006003388A2 (en) | 2004-06-30 | 2006-01-12 | Domantis Limited | Compositions and methods for treating inflammatory disorders |
WO2006030220A1 (en) | 2004-09-17 | 2006-03-23 | Domantis Limited | Compositions monovalent for cd40l binding and methods of use |
WO2012049570A1 (en) | 2010-10-11 | 2012-04-19 | Panima Pharmaceuticals Ag | Human anti-tau antibodies |
US20140068797A1 (en) | 2012-05-25 | 2014-03-06 | University Of Vienna | Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription |
US8697359B1 (en) | 2012-12-12 | 2014-04-15 | The Broad Institute, Inc. | CRISPR-Cas systems and methods for altering expression of gene products |
WO2014096321A1 (en) | 2012-12-21 | 2014-06-26 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Antibodies specific to tau phosphorylated at serine 422 and uses for the treatment and diagnosis of tauopathies |
WO2015004163A1 (en) | 2013-07-08 | 2015-01-15 | Adx Neurosciences | Anti-tau monoclonal antibodies |
WO2015200806A2 (en) | 2014-06-27 | 2015-12-30 | C2N Diagnostics Llc | Humanized anti-tau antibodies |
WO2016112078A2 (en) | 2015-01-08 | 2016-07-14 | Janssen Biotech, Inc. | Anti-phf-tau antibodies and their uses |
WO2018152359A1 (en) | 2017-02-17 | 2018-08-23 | Denali Therapeutics Inc. | Anti-tau antibodies and methods of use thereof |
WO2020120644A1 (en) | 2018-12-13 | 2020-06-18 | INSERM (Institut National de la Santé et de la Recherche Médicale) | New anti tau svqivykpv epitope single domain antibody |
WO2020193520A1 (en) | 2019-03-25 | 2020-10-01 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Treatment of taupathy disorders by targeting new tau species |
WO2021010712A1 (en) | 2019-07-15 | 2021-01-21 | 주식회사 아델 | Anti-tau antibody and use of same |
Non-Patent Citations (74)
Title |
---|
"Uniprot", Database accession no. P 1063 6-7 |
AGNEW CHEM INTL. ED. ENGL., vol. 33, 1994, pages 183 - 186 |
ASADA-UTSUGI, M.UEMURA, K.AYAKI, T.M, T.U.MINAMIYAMA, S.HIKIAMI, R.MORIMURA, T.SHODAI, A.UEKI, T.TAKAHASHI, R. ET AL.: "Failure of DNA double-strand break repair by tau mediates alzheimer's disease pathology in vitro", COMMUN BIOL, vol. 5, 2022, pages 358 |
ATKINSON, J.BEZAK, E.KEMPSON, I: "Imaging DNA double-strand breaks - are we there yet?", NAT REV MOL CELL BIOL, vol. 23, 2022, pages 579 - 580 |
BASU, A.KRISHNAMURTHY, S: "Cellular responses to cisplatin-induced DNA damage", J NUCLEIC ACIDS, 2010 |
BENNARDO, N.CHENG, A.HUANG, N.STARK, J.M.: "Alternative-nhej is a mechanistically distinct pathway of mammalian chromosome break repair", PLOS GENET, vol. 4, 2008, XP002680904, DOI: 10.1371/JOURNAL.PGEN.1000110 |
BEUCHER, A.BIRRAUX, J.TCHOUANDONG, L.BARTON, O.SHIBATA, ACONRAD, S.GOODARZI, A.A.KREMPLER, A.JEGGO, P.A.LOBRICH, M.: "Atm and artemis promote homologous recombination of radiation-induced DNA double-strand breaks in g2", EMBO J2009, vol. 28, pages 3413 - 3427 |
CECCALDI, R.RONDINELLI, B.D'ANDREA, A.D.: "Repair pathway choices and consequences at the double-strand break", TRENDS CELL BIOL, vol. 26, 2016, pages 52 - 64, XP029373553, DOI: 10.1016/j.tcb.2015.07.009 |
CHAPMAN, J.RSOSSICK, A.J.BOULTON, S.J.JACKSON, S.P: "Brcal-associated exclusion of 53bp1 from DNA damage sites underlies temporal control of DNA repair", J CELL SCI, vol. 125, 2012, pages 3529 - 3534, XP055593608, DOI: 10.1242/jcs.105353 |
CHAUDERLIER, A.GILLES, M.SPOLCOVA, A.CAILLIEREZ, R.CHWASTYNIAK, M.KRESS, M.DROBECQ, H.BONNEFOY, E.PINET, FWEIL, D. ET AL.: "Tau/ddx6 interaction increases microrna activity", BIOCHIM BIOPHYS ACTA GENE REGUL MECH, vol. 1861, 2018, pages 762 - 772, XP085427612, DOI: 10.1016/j.bbagrm.2018.06.006 |
CHAUDHARY, A.R.BERGER, F.BERGER, C.L.HENDRICKS, A.G.: "Tau directs intracellular trafficking by regulating the forces exerted by kinesin and dynein teams", TRAFFIC, vol. 19, 2018, pages 111 - 121 |
CLOUAIRE, T.LEGUBE, G: "A snapshot on the cis chromatin response to DNA double-strand breaks", TRENDS GENET, vol. 35, 2019, pages 330 - 345 |
COLAS ET AL., NATURE, vol. 380, 1996, pages 548 - 50 |
COLNAGHI, L.RONDELLI, D.MUZI-FALCONI, M.SERTIC, S: "Tau and DNA damage in neurodegeneration", BRAIN SCI, vol. 10, 2020 |
DELOBEL, P.FLAMENT, S.HAMDANE, M.JAKES, R.ROUSSEAU, A.DELACOURTE, AVILAIN, J.PGOEDERT, M.BUEE, L: "Functional characterization of ftdp-17 tau gene mutations through their effects on xenopus oocyte maturation", JBIOL CHEM, vol. 277, 2002, pages 9199 - 9205 |
DERISBOURG, M.LEGHAY, C.CHIAPPETTA, G.FERNANDEZ-GOMEZ, F.J.LAURENT, C.DEMEYER, D.CARRIER, SBUEE-SCHERRER, V.BLUM, D.VINH, J. ET AL: "Role of the tau n-terminal region in microtubule stabilization revealed by new endogenous truncated forms", SCI REP, vol. 5, 2015, pages 9659, XP055395382, DOI: 10.1038/srep09659 |
DICARLO ET AL., NUCLEIC ACIDS RES., vol. 41, 2013, pages 4336 - 4343 |
DIEZ, L.WEGMANN, S: "Nuclear transport deficits in tau-related neurodegenerative diseases", FRONT NEUROL, vol. 11, 2020, pages 1056 |
E. W. MARTIN: "Remington's Pharmaceutical-Sciences, Sixteenth", 1980, MACK PUBLISHING CO., EASTON |
FABRE ET AL., PLOS NEGL. TROP. DIS., vol. 8, 2014, pages e2671 |
FROST, B.HEMBERG, M.LEWIS, J.FEANY, M.B: "Tau promotes neurodegeneration through global chromatin relaxation", NAT NEUROSCI, vol. 17, 2014, pages 357 - 366 |
GALAS, M.C.BONNEFOY, E.BUEE, LLEFEBVRE, B: "Emerging connections between tau and nucleic acids", ADV EXP MEDBIOL, vol. 1184, 2019, pages 135 - 143 |
GIANNAKAKOU, P.NAKANO, MNICOLAOU, K.C.O'BRATE, A.YU, J.BLAGOSKLONNY, M.V.GREBER, U.F.FOJO, T: "Enhanced microtubule-dependent trafficking and p53 nuclear accumulation by suppression of microtubule dynamics", PROC NATL ACAD SCI USA, vol. 99, 2002, pages 10855 - 10860 |
GIANNAKAKOU, P.SACKETT, D.L.WARD, Y.WEBSTER, K.R.BLAGOSKLONNY, M.V.FOJO, T: "P53 is associated with cellular microtubules and is transported to the nucleus by dynein", NAT CELL BIOL, vol. 2, 2000, pages 709 - 717 |
GONZALEZ-SUAREZ, I.REDWOOD, A.B.GROTSKY, D.A.NEUMANN, M.A.CHENG, E.H.STEWART, C.L.DUSSO, A.GONZALO, S: "A new pathway that regulates 53bp 1 stability implicates cathepsin 1 and vitamin d in DNA repair", EMBO J2011, vol. 30, pages 3383 - 3396 |
GRATZ ET AL., GENETICS, 2014 |
GUO ET AL., DEVELOPMENT, vol. 141, 2014, pages 707 - 714 |
HAI ET AL., CELL RES, 2014 |
HEDNA RAYANE ET AL: "Tau Protein as Therapeutic Target for Cancer? Focus on Glioblastoma", CANCERS, vol. 14, no. 21, 1 November 2022 (2022-11-01), pages 5386, XP093042070, DOI: 10.3390/cancers14215386 * |
HEDNA, RKOVACIC, H.PAGANO, A.PEYROT, V.ROBIN, M.DEVRED, F.BREUZARD, G: "Tau protein as therapeutic target for cancer? Focus on glioblastoma", CANCERS (BASEL, 2022, pages 14 |
HER, J.BUNTING, S.F: "How cells ensure correct repair of DNA double-strand breaks", J BIOL CHEM, vol. 293, 2018, pages 10502 - 10511, XP055830556, DOI: 10.1074/jbc.TM118.000371 |
HWANG ET AL., PLOS ONE, vol. 8, 2013, pages e68708 |
JADHAV ET AL., ACTA NEUROPATHOLOGICA COMMUNICATIONS, vol. 7, 2019, pages 22 |
JAYASENA S.D., CLIN. CHEM., vol. 45, no. 9, 1999, pages 1628 - 50 |
KHANNA, K.K.JACKSON, S.P: "DNA double-strand breaks: Signaling, repair and the cancer connection", NAT GENET, vol. 27, 2001, pages 247 - 254 |
KURIHARA, M.MANO, T.SAITO, Y.MURAYAMA, S.TODA, T.IWATA, A: "Colocalization of brcal with tau aggregates in human tauopathies", BRAIN SCI, vol. 10, 2019 |
LESTER, E.PARKER, R: "The tau of nuclear-cytoplasmic transport", NEURON, vol. 99, 2018, pages 869 - 871, XP085465586, DOI: 10.1016/j.neuron.2018.08.026 |
LI, Q.MARTINEZ, J.D.: "P53 is transported into the nucleus via an hsfl-dependent nuclear localization mechanism", MOL CARCINOG, vol. 50, 2011, pages 143 - 152 |
LOTTERSBERGER, F.KARSSEMEIJER, R.A.DIMITROVA, NDE LANGE, T: "53bp1 and the line complex promote microtubule-dependent dsb mobility and DNA repair", CELL, vol. 163, 2015, pages 880 - 893 |
LU M ET AL.: "Discovery of a Keapl-dependent peptide PROTAC to knockdown Tau", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 146, 2018, pages 251e259 |
MA ET AL., CELL RES., vol. 24, 2014, pages 122 - 125 |
MALI ET AL., SCIENCE, vol. 339, 2013, pages 823 - 826 |
MAO, Z.BOZZELLA, M.SELUANOV, A.GORBUNOVA, V: "Comparison of nonhomologous end joining and homologous recombination in human cells", DNA REPAIR (AMST), vol. 7, 2008, pages 1765 - 1771, XP025466774, DOI: 10.1016/j.dnarep.2008.06.018 |
MARTA SMOTER ET AL: "The role of Tau protein in resistance to paclitaxel", CANCER CHEMOTHERAPY AND PHARMACOLOGY, SPRINGER, BERLIN, DE, vol. 68, no. 3, 29 June 2011 (2011-06-29), pages 553 - 557, XP019944719, ISSN: 1432-0843, DOI: 10.1007/S00280-011-1696-7 * |
MASHIKO ET AL., DEV. GROWTH DIFFER, vol. 56, 2014, pages 122 - 129 |
MEKHAIL, K: "Corrigendum: Defining the damaged DNA mobility paradox as revealed by the study of telomeres, dsbs, microtubules and motors", FRONT GENET, vol. 9, 2018, pages 157 |
MEKHAIL, K: "Defining the damaged DNA mobility paradox as revealed by the study of telomeres, dsbs, microtubules and motors", FRONT GENET, vol. 9, 2018, pages 95 |
NAKAMURA, MKANEKO, S.DICKSON, D.W.KUSAKA, H: "Aberrant accumulation of brcal in alzheimer disease and other tauopathies", JNEUROPATHOL EXP NEUROL, vol. 79, 2020, pages 22 - 33 |
NIU ET AL., CELL, vol. 156, 2014, pages 836 - 843 |
OSHIDARI, RSTRECKER, J.CHUNG, D.K.C.ABRAHAM, K.J.CHAN, J.N.Y.DAMAREN, C.J.MEKHAIL, K: "Nuclear microtubule filaments mediate non-linear directional motion of chromatin and promote DNA repair", NAT COMMUN, vol. 9, 2018, pages 2567 |
PAONESSA, FEVANS, L.D.SOLANKI, R.LARRIEU, D.WRAY, S.HARDY, J.JACKSON, S.P.LIVESEY, F.J.: "Microtubules deform the nuclear membrane and disrupt nucleocytoplasmic transport in tau-mediated frontotemporal dementia", CELL REP, vol. 26, 2019, pages 582 - 593 |
PAPIN SPAGANETTI P: "Emerging Evidences for an Implication of the Neurodegeneration-Associated Protein TAU", CANCER. BRAIN SCI, vol. 10, no. 11, 16 November 2020 (2020-11-16), pages 862 |
PAPIN, S.PAGANETTI, P: "Emerging evidences for an implication of the neurodegeneration-associated protein tau in cancer", BRAIN SCI, vol. 10, 2020 |
PIERCE, A.J.JOHNSON, R.D.THOMPSON, L.H.JASIN, M: "Xrcc3 promotes homology-directed repair of DNA damage in mammalian cells", GENES DEV, vol. 13, 1999, pages 2633 - 2638, XP001000711, DOI: 10.1101/gad.13.20.2633 |
PORUCHYNSKY, M.S.KOMLODI-PASZTOR, E.TROSTEL, S.WILKERSON, J.REGAIRAZ, M.POMMIER, Y.ZHANG, X.KUMAR MAITY, T.ROBEY, R.BUROTTO, M. ET: "Microtubule-targeting agents augment the toxicity of DNA-damaging agents by disrupting intracellular trafficking of DNA repair proteins", PROC NATL ACAD SCI USA, vol. 112, 2015, pages 1571 - 1576 |
QI, H.CANTRELLE, F.X.BENHELLI-MOKRANI, H.SMET-NOCCA, C.BUEE, L.LIPPENS, G.BONNEFOY, E.GALAS, M.C.LANDRIEU, I: "Nuclear magnetic resonance spectroscopy characterization of interaction of tau with DNA and its regulation by phosphorylation", BIOCHEMISTRY, vol. 54, 2015, pages 1525 - 1533 |
REYNOLDS, C. H. ET AL., J. NEUROCHEM., vol. 69, 1997, pages 191 - 198 |
RICO, T.GILLES, M.CHAUDERLIER, A.COMPTDAER, T.MAGNEZ, R.CHWASTYNIAK, M.DROBECQ, H.PINET, F.THURU, X.BUEE, L ET AL.: "Tau stabilizes chromatin compaction", FRONT CELL DEV BIOL, vol. 9, 2021, pages 740550 |
ROSSI, G.REDAELLI, V.PEREGO, P.FERRARI, R.GIACCONE, G.TAGLIAVINI, F: "Tau mutations as a novel risk factor for cancer-response", CANCER RES, vol. 78, 2018, pages 6525 |
ROUZIER ROMAN ET AL: "Microtubule-associated protein tau: A marker of paclitaxel sensitivity in breast cancer Departments of a Breast Medical Oncology, c Pathology, d Biostatistics and Applied Mathematics, j Radiation Oncology, and k Surgical Oncology, University of", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, 24 May 2005 (2005-05-24), pages 8315 - 8320, XP093042384, Retrieved from the Internet <URL:www.pnas.org/cgi/doi/10.1073/pnas.0408974102> [retrieved on 20230426] * |
SILVA, M.J.COSTA, P.DIAS, A.VALENTE, M.LOURO, H.BOAVIDA, M.G: "Comparative analysis of the mutagenic activity of oxaliplatin and cisplatin in the hprt gene of cho cells", ENVIRON MOLMUTAGEN, vol. 46, 2005, pages 104 - 115, XP071736183, DOI: 10.1002/em.20138 |
SMOTER M. ET AL., JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH, vol. 32, 2013, pages 25 |
SOTIROPOULOS, I.GALAS, M.C.SILVA, J.M.SKOULAKIS, E.WEGMANN, S.MAINA, M.B.BLUM, D.SAYAS, C.L.MANDELKOW, E.M.MANDELKOW, E. ET AL.: "Atypical, non-standard functions of the microtubule associated tau protein", ACTA NEUROPATHOL COMMUN, vol. 5, 2017, pages 91 |
SULTAN, A.NESSLANY, F.VIOLET, M.BEGARD, S.LOYENS, A.TALAHARI, S.MANSUROGLU, Z.MARZIN, D.SERGEANT, N.HUMEZ, S ET AL.: "Nuclear tau, a key player in neuronal DNA protection", JBIOL CHEM, vol. 286, 2011, pages 4566 - 4575 |
SUOMALAINEN, M.NAKANO, M.Y.KELLER, S.BOUCKE, K.STIDWILL, R.P.GREBER, U.F.: "Microtubule-dependent plus- and minus end-directed motilities are competing processes for nuclear targeting of adenovirus", J CELL BIOL, vol. 144, 1999, pages 657 - 672 |
TRENNER, A.; SARTORI, A.A: "Harnessing DNA double-strand break repair for cancer treatment", FRONT ONCOL, vol. 9, 2019, pages 1388 |
TUERK CGOLD L, SCIENCE, vol. 249, no. 4968, 1990, pages 505 - 10 |
VIOLET, M.CHAUDERLIER, A.DELATTRE, L.TARDIVEL, M.CHOUALA, M.S.SULTAN, A.MARCINIAK, E.HUMEZ, S.BINDER, L.KAYED, R. ET AL.: "Prefibrillar tau oligomers alter the nucleic acid protective function of tau in hippocampal neurons in vivo", NEUROBIOL DIS, vol. 82, 2015, pages 540 - 551 |
VIOLET, M.DELATTRE, L.TARDIVEL, MSULTAN, A.CHAUDERLIER, A.CAILLIEREZ, R.TALAHARI, S.NESSLANY, F.LEFEBVRE, BBONNEFOY, E. ET AL.: "A major role for tau in neuronal DNA and rna protection in vivo under physiological and hyperthermic conditions", FRONT CELL NEUROSCI, vol. 8, no. 84, 2014 |
WANG Q ET AL., PATHOL. ONCOL. RES, vol. 19, 2013, pages 429 - 435 |
WEI, Y.QU, M.H.WANG, X.S.CHEN, L.WANG, D.L.LIU, Y.HUA, Q.HE, R.Q: "Binding to the minor groove of the double-strand, tau protein prevents DNA from damage by peroxidation", PLOS ONE, vol. 3, 2008, pages e2600 |
WEINGARTEN, M.D.LOCKWOOD, A.H.HWO, S.Y.KIRSCHNER, M.W: "A protein factor essential for microtubule assembly", PROC NATL ACAD SCI USA, vol. 72, 1975, pages 1858 - 1862 |
YANG ET AL., J. MOL. CELL BIOL., vol. 6, 2014, pages 97 - 99 |
ZETSCHE ET AL.: "Cpfl is a Single RNA-guided Endonuclease of a Class 2 CRISPR-Cas System", CELL, vol. 163, 2015, pages 1 - 13 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Gorecki et al. | Discovery of ATR kinase inhibitor berzosertib (VX-970, M6620): Clinical candidate for cancer therapy | |
Liu et al. | Triple negative breast cancer therapy with CDK1 siRNA delivered by cationic lipid assisted PEG-PLA nanoparticles | |
Prete et al. | Pericytes elicit resistance to vemurafenib and sorafenib therapy in thyroid carcinoma via the TSP-1/TGFβ1 axis | |
JP7090933B2 (en) | How to Treat Cancers with Hemizygous Loss of TP53 | |
KR20200040763A (en) | Targeting kinase for the treatment of cancer metastasis | |
US20090192118A1 (en) | Tak1-d mediated induction of cell death in human cancer cells by specific sequence short double-stranded rnas | |
Berardinelli et al. | G-quadruplex ligand RHPS4 radiosensitizes glioblastoma xenograft in vivo through a differential targeting of bulky differentiated-and stem-cancer cells | |
Zeng et al. | CDK5 activates hippo signaling to confer resistance to radiation therapy via upregulating TAZ in lung cancer | |
US20190076464A1 (en) | Cytidine deaminase inhibitors for the treatment of pancreatic cancer | |
US20210171958A1 (en) | Methods of treating cancer | |
WO2020056188A1 (en) | Combination of parp inhibitor and brd4 inhibitor for the treatment of cancer | |
Jebelli et al. | Recent developments in targeting genes and pathways by RNAi‐based approaches in colorectal cancer | |
US20220193205A1 (en) | Methods of treating cancer | |
US10214743B2 (en) | Colorectal cancer drug, and method for predicting prognosis of colorectal cancer patient | |
US9623109B2 (en) | Methods of killing cells and use of same in prevention and treatment of cancer | |
WO2018078083A1 (en) | New method for treating multiple myeloma | |
Stoczynska-Fidelus et al. | Role of senescence in tumorigenesis and anticancer therapy | |
WO2016066671A1 (en) | Method for treating resistant cancers using progastrin inhibitors | |
EP3538140A1 (en) | Methods and pharmaceutical compositions for modulating stem cells proliferation or differentiation | |
WO2024133723A1 (en) | Methods for decreasing therapeutic acquired resistance to chemotherapy and/or radiotherapy | |
WO2023076880A1 (en) | Foxo1-targeted therapy for the treatment of cancer | |
CA2980611A1 (en) | A method of treating neoplasias | |
EP3963109A1 (en) | Methods and compositions for treating melanoma | |
WO2022084531A1 (en) | Methods and compositions for treating glioma | |
US20240165094A1 (en) | Methods and compositions for treating melanoma |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 23837319 Country of ref document: EP Kind code of ref document: A1 |