WO2021013942A1 - Use of myeloperoxidase inhibitors for the treatment of cardiovascular diseases in patients suffering from myeloproliferative neoplasms - Google Patents
Use of myeloperoxidase inhibitors for the treatment of cardiovascular diseases in patients suffering from myeloproliferative neoplasms Download PDFInfo
- Publication number
- WO2021013942A1 WO2021013942A1 PCT/EP2020/070827 EP2020070827W WO2021013942A1 WO 2021013942 A1 WO2021013942 A1 WO 2021013942A1 EP 2020070827 W EP2020070827 W EP 2020070827W WO 2021013942 A1 WO2021013942 A1 WO 2021013942A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- jak2
- microvesicles
- mice
- thioxo
- tetrahydro
- Prior art date
Links
- 201000007224 Myeloproliferative neoplasm Diseases 0.000 title claims abstract description 68
- 208000024172 Cardiovascular disease Diseases 0.000 title claims abstract description 9
- 229940122159 Myeloperoxidase inhibitor Drugs 0.000 title claims description 14
- 238000011282 treatment Methods 0.000 title description 20
- 210000003743 erythrocyte Anatomy 0.000 claims abstract description 85
- 230000007211 cardiovascular event Effects 0.000 claims abstract description 31
- 210000004369 blood Anatomy 0.000 claims description 34
- 239000008280 blood Substances 0.000 claims description 34
- 238000000034 method Methods 0.000 claims description 29
- 230000035772 mutation Effects 0.000 claims description 22
- 208000032027 Essential Thrombocythemia Diseases 0.000 claims description 13
- 208000017733 acquired polycythemia vera Diseases 0.000 claims description 13
- 208000037244 polycythemia vera Diseases 0.000 claims description 13
- 101000997832 Homo sapiens Tyrosine-protein kinase JAK2 Proteins 0.000 claims description 12
- 102100033444 Tyrosine-protein kinase JAK2 Human genes 0.000 claims description 11
- 229940121710 HMGCoA reductase inhibitor Drugs 0.000 claims description 10
- 206010028537 myelofibrosis Diseases 0.000 claims description 10
- 208000003476 primary myelofibrosis Diseases 0.000 claims description 7
- 241000699670 Mus sp. Species 0.000 abstract description 123
- 102000003896 Myeloperoxidases Human genes 0.000 abstract description 48
- 108090000235 Myeloperoxidases Proteins 0.000 abstract description 48
- 230000001965 increasing effect Effects 0.000 abstract description 48
- 230000008602 contraction Effects 0.000 abstract description 38
- 230000003511 endothelial effect Effects 0.000 abstract description 36
- 230000000694 effects Effects 0.000 abstract description 27
- 230000014509 gene expression Effects 0.000 abstract description 25
- 230000036542 oxidative stress Effects 0.000 abstract description 25
- 239000003112 inhibitor Substances 0.000 abstract description 15
- 102200087780 rs77375493 Human genes 0.000 abstract description 11
- 101150009057 JAK2 gene Proteins 0.000 abstract description 6
- 230000034994 death Effects 0.000 abstract description 4
- 231100000517 death Toxicity 0.000 abstract description 4
- 230000001404 mediated effect Effects 0.000 abstract description 4
- 230000003389 potentiating effect Effects 0.000 abstract description 3
- 230000003190 augmentative effect Effects 0.000 abstract description 2
- 230000010417 nitric oxide pathway Effects 0.000 abstract description 2
- MWUXSHHQAYIFBG-UHFFFAOYSA-N Nitric oxide Chemical compound O=[N] MWUXSHHQAYIFBG-UHFFFAOYSA-N 0.000 description 38
- 210000002889 endothelial cell Anatomy 0.000 description 38
- 210000004027 cell Anatomy 0.000 description 28
- 229960001802 phenylephrine Drugs 0.000 description 28
- SONNWYBIRXJNDC-VIFPVBQESA-N phenylephrine Chemical compound CNC[C@H](O)C1=CC=CC(O)=C1 SONNWYBIRXJNDC-VIFPVBQESA-N 0.000 description 28
- 238000002474 experimental method Methods 0.000 description 25
- 231100000673 dose–response relationship Toxicity 0.000 description 24
- 230000004044 response Effects 0.000 description 24
- 239000003981 vehicle Substances 0.000 description 24
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 23
- 108090000623 proteins and genes Proteins 0.000 description 23
- 210000000709 aorta Anatomy 0.000 description 22
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 22
- 230000001186 cumulative effect Effects 0.000 description 19
- RYMZZMVNJRMUDD-UHFFFAOYSA-N SJ000286063 Natural products C12C(OC(=O)C(C)(C)CC)CC(C)C=C2C=CC(C)C1CCC1CC(O)CC(=O)O1 RYMZZMVNJRMUDD-UHFFFAOYSA-N 0.000 description 16
- 235000018102 proteins Nutrition 0.000 description 16
- 102000004169 proteins and genes Human genes 0.000 description 16
- 239000003642 reactive oxygen metabolite Substances 0.000 description 16
- 239000000523 sample Substances 0.000 description 16
- 229960002855 simvastatin Drugs 0.000 description 16
- RYMZZMVNJRMUDD-HGQWONQESA-N simvastatin Chemical compound C([C@H]1[C@@H](C)C=CC2=C[C@H](C)C[C@@H]([C@H]12)OC(=O)C(C)(C)CC)C[C@@H]1C[C@@H](O)CC(=O)O1 RYMZZMVNJRMUDD-HGQWONQESA-N 0.000 description 16
- 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 15
- 208000010125 myocardial infarction Diseases 0.000 description 15
- 239000002953 phosphate buffered saline Substances 0.000 description 15
- -1 3-chloropyridin-2-yl Chemical group 0.000 description 14
- 210000002381 plasma Anatomy 0.000 description 14
- 238000011002 quantification Methods 0.000 description 14
- 210000000952 spleen Anatomy 0.000 description 14
- 238000004820 blood count Methods 0.000 description 13
- 239000000203 mixture Substances 0.000 description 13
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 description 13
- HFNKQEVNSGCOJV-OAHLLOKOSA-N ruxolitinib Chemical compound C1([C@@H](CC#N)N2N=CC(=C2)C=2C=3C=CNC=3N=CN=2)CCCC1 HFNKQEVNSGCOJV-OAHLLOKOSA-N 0.000 description 13
- PWKSKIMOESPYIA-BYPYZUCNSA-N L-N-acetyl-Cysteine Chemical compound CC(=O)N[C@@H](CS)C(O)=O PWKSKIMOESPYIA-BYPYZUCNSA-N 0.000 description 12
- 239000002144 L01XE18 - Ruxolitinib Substances 0.000 description 12
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 12
- 210000003038 endothelium Anatomy 0.000 description 12
- 229960000215 ruxolitinib Drugs 0.000 description 12
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 11
- 239000011859 microparticle Substances 0.000 description 11
- 108090000765 processed proteins & peptides Proteins 0.000 description 11
- VSNHCAURESNICA-UHFFFAOYSA-N Hydroxyurea Chemical compound NC(=O)NO VSNHCAURESNICA-UHFFFAOYSA-N 0.000 description 10
- 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 10
- 238000004458 analytical method Methods 0.000 description 10
- 230000003394 haemopoietic effect Effects 0.000 description 10
- 229960001330 hydroxycarbamide Drugs 0.000 description 10
- 238000010172 mouse model Methods 0.000 description 10
- 230000002792 vascular Effects 0.000 description 10
- 108090000672 Annexin A5 Proteins 0.000 description 9
- 102000004121 Annexin A5 Human genes 0.000 description 9
- 102100038055 Glutathione S-transferase theta-1 Human genes 0.000 description 9
- 239000000872 buffer Substances 0.000 description 9
- 238000005119 centrifugation Methods 0.000 description 9
- 150000001875 compounds Chemical class 0.000 description 9
- 239000003814 drug Substances 0.000 description 9
- 108010027853 glutathione S-transferase T1 Proteins 0.000 description 9
- 238000001727 in vivo Methods 0.000 description 9
- 230000005764 inhibitory process Effects 0.000 description 9
- 239000005526 vasoconstrictor agent Substances 0.000 description 9
- 241001465754 Metazoa Species 0.000 description 8
- 210000001105 femoral artery Anatomy 0.000 description 8
- 210000000265 leukocyte Anatomy 0.000 description 8
- 230000037361 pathway Effects 0.000 description 8
- 230000035945 sensitivity Effects 0.000 description 8
- 239000006228 supernatant Substances 0.000 description 8
- 238000012360 testing method Methods 0.000 description 8
- 239000013598 vector Substances 0.000 description 8
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 7
- 208000004476 Acute Coronary Syndrome Diseases 0.000 description 7
- 238000000585 Mann–Whitney U test Methods 0.000 description 7
- 241000699666 Mus <mouse, genus> Species 0.000 description 7
- 102100022698 NACHT, LRR and PYD domains-containing protein 1 Human genes 0.000 description 7
- 230000037396 body weight Effects 0.000 description 7
- 229940079593 drug Drugs 0.000 description 7
- 239000007924 injection Substances 0.000 description 7
- 238000002347 injection Methods 0.000 description 7
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 7
- WCUXLLCKKVVCTQ-UHFFFAOYSA-M potassium chloride Inorganic materials [Cl-].[K+] WCUXLLCKKVVCTQ-UHFFFAOYSA-M 0.000 description 7
- 230000002829 reductive effect Effects 0.000 description 7
- 239000000243 solution Substances 0.000 description 7
- 238000011740 C57BL/6 mouse Methods 0.000 description 6
- 238000000540 analysis of variance Methods 0.000 description 6
- 230000003078 antioxidant effect Effects 0.000 description 6
- 239000000074 antisense oligonucleotide Substances 0.000 description 6
- 238000012230 antisense oligonucleotides Methods 0.000 description 6
- 230000003247 decreasing effect Effects 0.000 description 6
- 208000035475 disorder Diseases 0.000 description 6
- 229940090044 injection Drugs 0.000 description 6
- 238000004949 mass spectrometry Methods 0.000 description 6
- BDAGIHXWWSANSR-UHFFFAOYSA-N methanoic acid Natural products OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 6
- 210000000056 organ Anatomy 0.000 description 6
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 6
- 102000004196 processed proteins & peptides Human genes 0.000 description 6
- 230000009257 reactivity Effects 0.000 description 6
- 230000001732 thrombotic effect Effects 0.000 description 6
- 206010002091 Anaesthesia Diseases 0.000 description 5
- 102100025621 Cytochrome b-245 heavy chain Human genes 0.000 description 5
- 102000004190 Enzymes Human genes 0.000 description 5
- 108090000790 Enzymes Proteins 0.000 description 5
- PIWKPBJCKXDKJR-UHFFFAOYSA-N Isoflurane Chemical compound FC(F)OC(Cl)C(F)(F)F PIWKPBJCKXDKJR-UHFFFAOYSA-N 0.000 description 5
- 229960004308 acetylcysteine Drugs 0.000 description 5
- 239000004480 active ingredient Substances 0.000 description 5
- 238000001949 anaesthesia Methods 0.000 description 5
- 230000037005 anaesthesia Effects 0.000 description 5
- 230000008859 change Effects 0.000 description 5
- 238000006243 chemical reaction Methods 0.000 description 5
- 201000010099 disease Diseases 0.000 description 5
- 229940088598 enzyme Drugs 0.000 description 5
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 5
- 150000003278 haem Chemical class 0.000 description 5
- 239000002471 hydroxymethylglutaryl coenzyme A reductase inhibitor Substances 0.000 description 5
- 238000011534 incubation Methods 0.000 description 5
- 150000002500 ions Chemical class 0.000 description 5
- 229960002725 isoflurane Drugs 0.000 description 5
- 230000007246 mechanism Effects 0.000 description 5
- 239000008188 pellet Substances 0.000 description 5
- 230000009467 reduction Effects 0.000 description 5
- 239000011780 sodium chloride Substances 0.000 description 5
- 229960001603 tamoxifen Drugs 0.000 description 5
- 230000001225 therapeutic effect Effects 0.000 description 5
- 238000001262 western blot Methods 0.000 description 5
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 4
- 108090000994 Catalytic RNA Proteins 0.000 description 4
- 102000053642 Catalytic RNA Human genes 0.000 description 4
- 201000000057 Coronary Stenosis Diseases 0.000 description 4
- 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 4
- 241000699660 Mus musculus Species 0.000 description 4
- 102000008299 Nitric Oxide Synthase Human genes 0.000 description 4
- 108010021487 Nitric Oxide Synthase Proteins 0.000 description 4
- DTQVDTLACAAQTR-UHFFFAOYSA-M Trifluoroacetate Chemical compound [O-]C(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-M 0.000 description 4
- 206010047249 Venous thrombosis Diseases 0.000 description 4
- 241000700605 Viruses Species 0.000 description 4
- OIPILFWXSMYKGL-UHFFFAOYSA-N acetylcholine Chemical compound CC(=O)OCC[N+](C)(C)C OIPILFWXSMYKGL-UHFFFAOYSA-N 0.000 description 4
- 229960004373 acetylcholine Drugs 0.000 description 4
- 230000000747 cardiac effect Effects 0.000 description 4
- 230000015556 catabolic process Effects 0.000 description 4
- 230000001413 cellular effect Effects 0.000 description 4
- 239000003153 chemical reaction reagent Substances 0.000 description 4
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 4
- 239000007928 intraperitoneal injection Substances 0.000 description 4
- 108020004999 messenger RNA Proteins 0.000 description 4
- 150000007523 nucleic acids Chemical class 0.000 description 4
- 230000002947 procoagulating effect Effects 0.000 description 4
- 230000006798 recombination Effects 0.000 description 4
- 238000005215 recombination Methods 0.000 description 4
- 238000012552 review Methods 0.000 description 4
- 108091092562 ribozyme Proteins 0.000 description 4
- 238000011830 transgenic mouse model Methods 0.000 description 4
- RSPDBEVKURKEII-UHFFFAOYSA-N 3-(oxolan-2-ylmethyl)-2-sulfanylidene-7h-purin-6-one Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CC1CCCO1 RSPDBEVKURKEII-UHFFFAOYSA-N 0.000 description 3
- 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 3
- OSWFIVFLDKOXQC-UHFFFAOYSA-N 4-(3-methoxyphenyl)aniline Chemical compound COC1=CC=CC(C=2C=CC(N)=CC=2)=C1 OSWFIVFLDKOXQC-UHFFFAOYSA-N 0.000 description 3
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 3
- 102400000345 Angiotensin-2 Human genes 0.000 description 3
- 101800000733 Angiotensin-2 Proteins 0.000 description 3
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 3
- 201000001320 Atherosclerosis Diseases 0.000 description 3
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- 206010048554 Endothelial dysfunction Diseases 0.000 description 3
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 3
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 3
- CZGUSIXMZVURDU-JZXHSEFVSA-N Ile(5)-angiotensin II Chemical compound C([C@@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC=1C=CC=CC=1)C([O-])=O)NC(=O)[C@@H](NC(=O)[C@H](CCCNC(N)=[NH2+])NC(=O)[C@@H]([NH3+])CC([O-])=O)C(C)C)C1=CC=C(O)C=C1 CZGUSIXMZVURDU-JZXHSEFVSA-N 0.000 description 3
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 3
- KCWZGJVSDFYRIX-YFKPBYRVSA-N N(gamma)-nitro-L-arginine methyl ester Chemical compound COC(=O)[C@@H](N)CCCN=C(N)N[N+]([O-])=O KCWZGJVSDFYRIX-YFKPBYRVSA-N 0.000 description 3
- 108010082739 NADPH Oxidase 2 Proteins 0.000 description 3
- 206010028980 Neoplasm Diseases 0.000 description 3
- 108091034117 Oligonucleotide Proteins 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 239000012891 Ringer solution Substances 0.000 description 3
- 108091027967 Small hairpin RNA Proteins 0.000 description 3
- 108020004459 Small interfering RNA Proteins 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 208000007536 Thrombosis Diseases 0.000 description 3
- 208000007814 Unstable Angina Diseases 0.000 description 3
- 230000004913 activation Effects 0.000 description 3
- 229950006323 angiotensin ii Drugs 0.000 description 3
- 210000002376 aorta thoracic Anatomy 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- 230000004888 barrier function Effects 0.000 description 3
- 210000002798 bone marrow cell Anatomy 0.000 description 3
- 208000029078 coronary artery disease Diseases 0.000 description 3
- 238000006731 degradation reaction Methods 0.000 description 3
- 238000012217 deletion Methods 0.000 description 3
- 230000037430 deletion Effects 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 239000006185 dispersion Substances 0.000 description 3
- 230000008694 endothelial dysfunction Effects 0.000 description 3
- 230000007717 exclusion Effects 0.000 description 3
- 235000019253 formic acid Nutrition 0.000 description 3
- 238000009472 formulation Methods 0.000 description 3
- 230000006870 function Effects 0.000 description 3
- 230000002068 genetic effect Effects 0.000 description 3
- 239000004615 ingredient Substances 0.000 description 3
- 230000002401 inhibitory effect Effects 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 239000002609 medium Substances 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 208000031225 myocardial ischemia Diseases 0.000 description 3
- 239000002773 nucleotide Substances 0.000 description 3
- 125000003729 nucleotide group Chemical group 0.000 description 3
- 230000001590 oxidative effect Effects 0.000 description 3
- 229940036309 phenylephrine injection Drugs 0.000 description 3
- 239000001103 potassium chloride Substances 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 150000003839 salts Chemical class 0.000 description 3
- 239000001509 sodium citrate Substances 0.000 description 3
- 210000000130 stem cell Anatomy 0.000 description 3
- 238000003860 storage Methods 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 210000001519 tissue Anatomy 0.000 description 3
- 210000000689 upper leg Anatomy 0.000 description 3
- 229940019333 vitamin k antagonists Drugs 0.000 description 3
- VHJLVAABSRFDPM-UHFFFAOYSA-N 1,4-dithiothreitol Chemical compound SCC(O)C(O)CS VHJLVAABSRFDPM-UHFFFAOYSA-N 0.000 description 2
- ZIIQCSMRQKCOCT-UHFFFAOYSA-N 2-acetamido-3-methyl-3-nitrososulfanylbutanoic acid Chemical compound CC(=O)NC(C(O)=O)C(C)(C)SN=O ZIIQCSMRQKCOCT-UHFFFAOYSA-N 0.000 description 2
- 125000004105 2-pyridyl group Chemical group N1=C([*])C([H])=C([H])C([H])=C1[H] 0.000 description 2
- XNHFAGRBSMMFKL-UHFFFAOYSA-N 2-sulfanylidene-3,7-dihydropurin-6-one Chemical compound O=C1NC(=S)NC2=C1NC=N2 XNHFAGRBSMMFKL-UHFFFAOYSA-N 0.000 description 2
- 108700028369 Alleles Proteins 0.000 description 2
- 206010002383 Angina Pectoris Diseases 0.000 description 2
- 200000000007 Arterial disease Diseases 0.000 description 2
- 206010003175 Arterial spasm Diseases 0.000 description 2
- 208000037260 Atherosclerotic Plaque Diseases 0.000 description 2
- 208000007257 Budd-Chiari syndrome Diseases 0.000 description 2
- 101710186553 Cytochrome b-245 heavy chain Proteins 0.000 description 2
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 2
- 206010016654 Fibrosis Diseases 0.000 description 2
- 108010024636 Glutathione Proteins 0.000 description 2
- 239000012981 Hank's balanced salt solution Substances 0.000 description 2
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 2
- 206010020751 Hypersensitivity Diseases 0.000 description 2
- 206010049694 Left Ventricular Dysfunction Diseases 0.000 description 2
- 101150082766 MPO gene Proteins 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- 241001529936 Murinae Species 0.000 description 2
- 229930040373 Paraformaldehyde Natural products 0.000 description 2
- 229920002556 Polyethylene Glycol 300 Polymers 0.000 description 2
- 208000007718 Stable Angina Diseases 0.000 description 2
- 108090000190 Thrombin Proteins 0.000 description 2
- 108090000631 Trypsin Proteins 0.000 description 2
- 102000004142 Trypsin Human genes 0.000 description 2
- 230000002159 abnormal effect Effects 0.000 description 2
- JUGOREOARAHOCO-UHFFFAOYSA-M acetylcholine chloride Chemical compound [Cl-].CC(=O)OCC[N+](C)(C)C JUGOREOARAHOCO-UHFFFAOYSA-M 0.000 description 2
- 229960004266 acetylcholine chloride Drugs 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 230000001154 acute effect Effects 0.000 description 2
- 208000026935 allergic disease Diseases 0.000 description 2
- 150000001413 amino acids Chemical group 0.000 description 2
- 238000002583 angiography Methods 0.000 description 2
- 239000003963 antioxidant agent Substances 0.000 description 2
- 235000006708 antioxidants Nutrition 0.000 description 2
- 210000001367 artery Anatomy 0.000 description 2
- 230000004071 biological effect Effects 0.000 description 2
- 210000000601 blood cell Anatomy 0.000 description 2
- 230000036471 bradycardia Effects 0.000 description 2
- 208000006218 bradycardia Diseases 0.000 description 2
- 239000011575 calcium Substances 0.000 description 2
- 229910052791 calcium Inorganic materials 0.000 description 2
- 210000000170 cell membrane Anatomy 0.000 description 2
- 210000003855 cell nucleus Anatomy 0.000 description 2
- 239000013522 chelant Substances 0.000 description 2
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 2
- 230000007882 cirrhosis Effects 0.000 description 2
- 208000019425 cirrhosis of liver Diseases 0.000 description 2
- 238000003776 cleavage reaction Methods 0.000 description 2
- 210000004351 coronary vessel Anatomy 0.000 description 2
- 239000013078 crystal Substances 0.000 description 2
- 238000001784 detoxification Methods 0.000 description 2
- 239000008121 dextrose Substances 0.000 description 2
- 238000002405 diagnostic procedure Methods 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 230000003828 downregulation Effects 0.000 description 2
- 238000002565 electrocardiography Methods 0.000 description 2
- 230000008753 endothelial function Effects 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 238000011067 equilibration Methods 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 230000009368 gene silencing by RNA Effects 0.000 description 2
- 239000011521 glass Substances 0.000 description 2
- 229960003180 glutathione Drugs 0.000 description 2
- 239000003102 growth factor Substances 0.000 description 2
- 230000011132 hemopoiesis Effects 0.000 description 2
- 102000049921 human JAK2 Human genes 0.000 description 2
- 230000009610 hypersensitivity Effects 0.000 description 2
- 230000001771 impaired effect Effects 0.000 description 2
- 230000006698 induction Effects 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 238000003780 insertion Methods 0.000 description 2
- 230000037431 insertion Effects 0.000 description 2
- 238000001990 intravenous administration Methods 0.000 description 2
- 238000010253 intravenous injection Methods 0.000 description 2
- NBQNWMBBSKPBAY-UHFFFAOYSA-N iodixanol Chemical compound IC=1C(C(=O)NCC(O)CO)=C(I)C(C(=O)NCC(O)CO)=C(I)C=1N(C(=O)C)CC(O)CN(C(C)=O)C1=C(I)C(C(=O)NCC(O)CO)=C(I)C(C(=O)NCC(O)CO)=C1I NBQNWMBBSKPBAY-UHFFFAOYSA-N 0.000 description 2
- PGHMRUGBZOYCAA-UHFFFAOYSA-N ionomycin Natural products O1C(CC(O)C(C)C(O)C(C)C=CCC(C)CC(C)C(O)=CC(=O)C(C)CC(C)CC(CCC(O)=O)C)CCC1(C)C1OC(C)(C(C)O)CC1 PGHMRUGBZOYCAA-UHFFFAOYSA-N 0.000 description 2
- 230000002427 irreversible effect Effects 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 230000002071 myeloproliferative effect Effects 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 210000000440 neutrophil Anatomy 0.000 description 2
- 108091027963 non-coding RNA Proteins 0.000 description 2
- 102000042567 non-coding RNA Human genes 0.000 description 2
- 238000010606 normalization Methods 0.000 description 2
- 102000039446 nucleic acids Human genes 0.000 description 2
- 108020004707 nucleic acids Proteins 0.000 description 2
- 230000000414 obstructive effect Effects 0.000 description 2
- 230000002018 overexpression Effects 0.000 description 2
- 229920002866 paraformaldehyde Polymers 0.000 description 2
- 230000001575 pathological effect Effects 0.000 description 2
- 230000007310 pathophysiology Effects 0.000 description 2
- 239000008194 pharmaceutical composition Substances 0.000 description 2
- 239000000546 pharmaceutical excipient Substances 0.000 description 2
- 230000036470 plasma concentration Effects 0.000 description 2
- 235000011164 potassium chloride Nutrition 0.000 description 2
- 238000011533 pre-incubation Methods 0.000 description 2
- 239000002243 precursor Substances 0.000 description 2
- 230000003244 pro-oxidative effect Effects 0.000 description 2
- 239000000047 product Substances 0.000 description 2
- 230000007017 scission Effects 0.000 description 2
- 238000000926 separation method Methods 0.000 description 2
- 208000007056 sickle cell anemia Diseases 0.000 description 2
- 239000002924 silencing RNA Substances 0.000 description 2
- 239000004055 small Interfering RNA Substances 0.000 description 2
- NLJMYIDDQXHKNR-UHFFFAOYSA-K sodium citrate Chemical compound O.O.[Na+].[Na+].[Na+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O NLJMYIDDQXHKNR-UHFFFAOYSA-K 0.000 description 2
- 235000011083 sodium citrates Nutrition 0.000 description 2
- 239000001488 sodium phosphate Substances 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 239000012453 solvate Substances 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 238000010561 standard procedure Methods 0.000 description 2
- 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 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 229960004072 thrombin Drugs 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- 230000014616 translation Effects 0.000 description 2
- 239000012588 trypsin Substances 0.000 description 2
- 241001430294 unidentified retrovirus Species 0.000 description 2
- 230000006492 vascular dysfunction Effects 0.000 description 2
- 230000004865 vascular response Effects 0.000 description 2
- 230000002227 vasoactive effect Effects 0.000 description 2
- 210000001631 vena cava inferior Anatomy 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- ZGGHKIMDNBDHJB-NRFPMOEYSA-M (3R,5S)-fluvastatin sodium Chemical compound [Na+].C12=CC=CC=C2N(C(C)C)C(\C=C\[C@@H](O)C[C@@H](O)CC([O-])=O)=C1C1=CC=C(F)C=C1 ZGGHKIMDNBDHJB-NRFPMOEYSA-M 0.000 description 1
- SEPPVOUBHWNCAW-FNORWQNLSA-N (E)-4-oxonon-2-enal Chemical compound CCCCCC(=O)\C=C\C=O SEPPVOUBHWNCAW-FNORWQNLSA-N 0.000 description 1
- TZCPCKNHXULUIY-RGULYWFUSA-N 1,2-distearoyl-sn-glycero-3-phosphoserine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP(O)(=O)OC[C@H](N)C(O)=O)OC(=O)CCCCCCCCCCCCCCCCC TZCPCKNHXULUIY-RGULYWFUSA-N 0.000 description 1
- SGKJJXCTYVZODW-UHFFFAOYSA-N 1,8-dimethyl-3-(2-methylpropyl)-6-sulfanylidene-7h-purin-2-one Chemical compound S=C1N(C)C(=O)N(CC(C)C)C2=C1NC(C)=N2 SGKJJXCTYVZODW-UHFFFAOYSA-N 0.000 description 1
- UZASYDVRUNALQB-UHFFFAOYSA-N 1-(1h-benzimidazol-2-ylmethyl)-2-sulfanylidene-5h-pyrrolo[3,2-d]pyrimidin-4-one Chemical compound C1=CC=C2NC(CN3C(=S)NC(C=4NC=CC=43)=O)=NC2=C1 UZASYDVRUNALQB-UHFFFAOYSA-N 0.000 description 1
- OGTLEZMDFONXBQ-UHFFFAOYSA-N 1-(2-methoxy-2-methylpropyl)-2-sulfanylidene-5h-pyrrolo[3,2-d]pyrimidin-4-one Chemical compound O=C1NC(=S)N(CC(C)(C)OC)C2=C1NC=C2 OGTLEZMDFONXBQ-UHFFFAOYSA-N 0.000 description 1
- XGZUURWEGKBYOK-UHFFFAOYSA-N 1-(piperidin-3-ylmethyl)-2-sulfanylidene-5h-pyrrolo[3,2-d]pyrimidin-4-one Chemical compound C1=2C=CNC=2C(=O)NC(=S)N1CC1CCCNC1 XGZUURWEGKBYOK-UHFFFAOYSA-N 0.000 description 1
- LJCUZCGIZWLJJY-UHFFFAOYSA-N 1-[(2-methoxyphenyl)methyl]-2-sulfanylidene-5h-pyrrolo[3,2-d]pyrimidin-4-one Chemical compound COC1=CC=CC=C1CN1C(=S)NC(=O)C2=C1C=CN2 LJCUZCGIZWLJJY-UHFFFAOYSA-N 0.000 description 1
- NVTFAAMTTOCCBM-UHFFFAOYSA-N 1-[(3-methoxyphenyl)methyl]-2-sulfanylidene-5h-pyrrolo[3,2-d]pyrimidin-4-one Chemical compound COC1=CC=CC(CN2C(NC(=O)C=3NC=CC=32)=S)=C1 NVTFAAMTTOCCBM-UHFFFAOYSA-N 0.000 description 1
- LHDQQAXJOKHNQC-UHFFFAOYSA-N 1-[(4,5-dimethyl-1h-benzimidazol-2-yl)methyl]-2-sulfanylidene-5h-pyrrolo[3,2-d]pyrimidin-4-one Chemical compound N=1C2=C(C)C(C)=CC=C2NC=1CN(C(NC1=O)=S)C2=C1NC=C2 LHDQQAXJOKHNQC-UHFFFAOYSA-N 0.000 description 1
- AWUPSBKDUQAAKD-UHFFFAOYSA-N 1-[(5-fluoro-1h-indol-2-yl)methyl]-2-sulfanylidene-5h-pyrrolo[3,2-d]pyrimidin-4-one Chemical compound C=1C2=CC(F)=CC=C2NC=1CN(C(NC1=O)=S)C2=C1NC=C2 AWUPSBKDUQAAKD-UHFFFAOYSA-N 0.000 description 1
- GXVILYVZTYQCRZ-UHFFFAOYSA-N 1-[(5-fluoro-1h-indol-3-yl)methyl]-2-sulfanylidene-5h-pyrrolo[3,2-d]pyrimidin-4-one Chemical compound C12=CC(F)=CC=C2NC=C1CN(C(NC1=O)=S)C2=C1NC=C2 GXVILYVZTYQCRZ-UHFFFAOYSA-N 0.000 description 1
- SWTXSGKQNZRCQK-UHFFFAOYSA-N 1-methyl-3-(2-methylpropyl)-2-sulfanylidene-7h-purin-6-one Chemical compound O=C1N(C)C(=S)N(CC(C)C)C2=C1NC=N2 SWTXSGKQNZRCQK-UHFFFAOYSA-N 0.000 description 1
- VSNHCAURESNICA-NJFSPNSNSA-N 1-oxidanylurea Chemical compound N[14C](=O)NO VSNHCAURESNICA-NJFSPNSNSA-N 0.000 description 1
- DNCYBUMDUBHIJZ-UHFFFAOYSA-N 1h-pyrimidin-6-one Chemical compound O=C1C=CN=CN1 DNCYBUMDUBHIJZ-UHFFFAOYSA-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
- MAQKUFVZHSCAGH-UHFFFAOYSA-N 3-(1h-pyrazol-5-ylmethyl)-2-sulfanylidene-7h-purin-6-one Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CC=1C=CNN=1 MAQKUFVZHSCAGH-UHFFFAOYSA-N 0.000 description 1
- GNFTZDOKVXKIBK-UHFFFAOYSA-N 3-(2-methoxyethoxy)benzohydrazide Chemical compound COCCOC1=CC=CC(C(=O)NN)=C1 GNFTZDOKVXKIBK-UHFFFAOYSA-N 0.000 description 1
- VXTADZREIUKIHH-UHFFFAOYSA-N 3-(2-methoxyethyl)-2-sulfanylidene-7h-purin-6-one Chemical compound O=C1NC(=S)N(CCOC)C2=C1NC=N2 VXTADZREIUKIHH-UHFFFAOYSA-N 0.000 description 1
- YWXBNZQTOMKSGH-UHFFFAOYSA-N 3-(2-methylbutyl)-6-sulfanylidene-7h-purin-2-one Chemical compound S=C1NC(=O)N(CC(C)CC)C2=C1NC=N2 YWXBNZQTOMKSGH-UHFFFAOYSA-N 0.000 description 1
- HWSAZERBFFRQGX-UHFFFAOYSA-N 3-(2-methylpropyl)-2-sulfanylidene-7h-purin-6-one Chemical compound O=C1NC(=S)N(CC(C)C)C2=C1NC=N2 HWSAZERBFFRQGX-UHFFFAOYSA-N 0.000 description 1
- KDGNPNTWFNHLSX-UHFFFAOYSA-N 3-(2-methylpropyl)-7h-purine-2,6-dithione Chemical compound S=C1NC(=S)N(CC(C)C)C2=C1NC=N2 KDGNPNTWFNHLSX-UHFFFAOYSA-N 0.000 description 1
- QYMIGFAAZFXYSQ-UHFFFAOYSA-N 3-(2-phenylethyl)-2-sulfanylidene-7h-purin-6-one Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CCC1=CC=CC=C1 QYMIGFAAZFXYSQ-UHFFFAOYSA-N 0.000 description 1
- BYEFLNKUKSUKMO-UHFFFAOYSA-N 3-(3-methoxypropyl)-2-sulfanylidene-7h-purin-6-one Chemical compound O=C1NC(=S)N(CCCOC)C2=C1NC=N2 BYEFLNKUKSUKMO-UHFFFAOYSA-N 0.000 description 1
- QTXZVSXYBGMMAU-UHFFFAOYSA-N 3-(cyclohexylmethyl)-2-sulfanylidene-7h-purin-6-one Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CC1CCCCC1 QTXZVSXYBGMMAU-UHFFFAOYSA-N 0.000 description 1
- KULKOCNEGWFYNO-UHFFFAOYSA-N 3-(cyclopropylmethyl)-2-sulfanylidene-7h-purin-6-one Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CC1CC1 KULKOCNEGWFYNO-UHFFFAOYSA-N 0.000 description 1
- XGZKFMGVJXEFAS-UHFFFAOYSA-N 3-(furan-2-ylmethyl)-2-sulfanylidene-7h-purin-6-one Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CC1=CC=CO1 XGZKFMGVJXEFAS-UHFFFAOYSA-N 0.000 description 1
- SCYILAUVHGNMOS-UHFFFAOYSA-N 3-[(1-methylindol-2-yl)methyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C1=CC=C2N(C)C(CN3C(NC(=O)C=4NC=NC=43)=S)=CC2=C1 SCYILAUVHGNMOS-UHFFFAOYSA-N 0.000 description 1
- GSEOEHAWXVFUCX-UHFFFAOYSA-N 3-[(3-propan-2-yl-1,2-oxazol-5-yl)methyl]-2-sulfanylidene-7h-purin-6-one Chemical compound O1N=C(C(C)C)C=C1CN1C(=S)NC(=O)C2=C1N=CN2 GSEOEHAWXVFUCX-UHFFFAOYSA-N 0.000 description 1
- GMNQNGTVFNOHLD-UHFFFAOYSA-N 3-[(4-fluorophenyl)methyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C1=CC(F)=CC=C1CN1C(=S)NC(=O)C2=C1N=CN2 GMNQNGTVFNOHLD-UHFFFAOYSA-N 0.000 description 1
- HZXVQEUOCFTRDT-UHFFFAOYSA-N 3-[(4-methoxyphenyl)methyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C1=CC(OC)=CC=C1CN1C(=S)NC(=O)C2=C1N=CN2 HZXVQEUOCFTRDT-UHFFFAOYSA-N 0.000 description 1
- SXNNLWVBOSOYAT-UHFFFAOYSA-N 3-[(5-chloro-1h-indol-3-yl)methyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C12=CC(Cl)=CC=C2NC=C1CN(C(NC1=O)=S)C2=C1NC=N2 SXNNLWVBOSOYAT-UHFFFAOYSA-N 0.000 description 1
- KHHYBTYRTYJWQL-UHFFFAOYSA-N 3-[2-(1h-benzimidazol-2-yl)ethyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C1=CC=C2NC(CCN3C(=S)NC(C=4NC=NC=43)=O)=NC2=C1 KHHYBTYRTYJWQL-UHFFFAOYSA-N 0.000 description 1
- XPJXEVNLVLNQRN-UHFFFAOYSA-N 3-[2-(1h-benzimidazol-2-ylmethylamino)propyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C1=CC=C2NC(CNC(CN3C(NC(=O)C=4NC=NC=43)=S)C)=NC2=C1 XPJXEVNLVLNQRN-UHFFFAOYSA-N 0.000 description 1
- ATPOLLKTGMVGBH-UHFFFAOYSA-N 3-[2-(pyridin-2-ylmethylamino)propyl]-2-sulfanylidene-7h-purin-6-one;2,2,2-trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F.C1=2N=CNC=2C(=O)NC(=S)N1CC(C)NCC1=CC=CC=N1 ATPOLLKTGMVGBH-UHFFFAOYSA-N 0.000 description 1
- RRQORCIDUZXJPT-UHFFFAOYSA-N 3-[2-(pyridin-3-ylmethylamino)propyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CC(C)NCC1=CC=CN=C1 RRQORCIDUZXJPT-UHFFFAOYSA-N 0.000 description 1
- YMEBQHCQKRPKCI-UHFFFAOYSA-N 3-[2-(pyridin-4-ylmethylamino)propyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CC(C)NCC1=CC=NC=C1 YMEBQHCQKRPKCI-UHFFFAOYSA-N 0.000 description 1
- SKAUBHPJNDJLBF-UHFFFAOYSA-N 3-[2-(quinolin-2-ylmethylamino)propyl]-2-sulfanylidene-7h-purin-6-one;2,2,2-trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F.C1=CC=CC2=NC(CNC(CN3C(NC(=O)C=4NC=NC=43)=S)C)=CC=C21 SKAUBHPJNDJLBF-UHFFFAOYSA-N 0.000 description 1
- DYKYDXGYDOPQCH-UHFFFAOYSA-N 3-[2-(quinolin-3-ylmethylamino)propyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C1=CC=CC2=CC(CNC(CN3C(NC(=O)C=4NC=NC=43)=S)C)=CN=C21 DYKYDXGYDOPQCH-UHFFFAOYSA-N 0.000 description 1
- CKBADCZMJZPWOE-UHFFFAOYSA-N 3-[2-[(1-methylindol-3-yl)methylamino]ethyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C12=CC=CC=C2N(C)C=C1CNCCN1C(=S)NC(=O)C2=C1N=CN2 CKBADCZMJZPWOE-UHFFFAOYSA-N 0.000 description 1
- RSEFPWXFDKRGNQ-UHFFFAOYSA-N 3-[2-[(1-methylindol-3-yl)methylamino]propyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C1=CC=C2C(CNC(CN3C(NC(=O)C=4NC=NC=43)=S)C)=CN(C)C2=C1 RSEFPWXFDKRGNQ-UHFFFAOYSA-N 0.000 description 1
- HJJAUEDOGPHQPS-UHFFFAOYSA-N 3-[2-[(1-methylpyrrol-2-yl)methylamino]propyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CC(C)NCC1=CC=CN1C HJJAUEDOGPHQPS-UHFFFAOYSA-N 0.000 description 1
- DSOXIQGGJIBKDX-UHFFFAOYSA-N 3-[2-[methyl(quinolin-4-ylmethyl)amino]ethyl]-2-sulfanylidene-7h-purin-6-one Chemical compound C1=CC=C2C(CN(CCN3C(NC(=O)C=4NC=NC=43)=S)C)=CC=NC2=C1 DSOXIQGGJIBKDX-UHFFFAOYSA-N 0.000 description 1
- RSPDBEVKURKEII-ZCFIWIBFSA-N 3-[[(2r)-oxolan-2-yl]methyl]-2-sulfanylidene-7h-purin-6-one Chemical group C1=2N=CNC=2C(=O)NC(=S)N1C[C@H]1CCCO1 RSPDBEVKURKEII-ZCFIWIBFSA-N 0.000 description 1
- PMCJHNWKVBFKTN-UHFFFAOYSA-N 3-butyl-2-sulfanylidene-7h-purin-6-one Chemical compound O=C1NC(=S)N(CCCC)C2=C1NC=N2 PMCJHNWKVBFKTN-UHFFFAOYSA-N 0.000 description 1
- 125000003349 3-pyridyl group Chemical group N1=C([H])C([*])=C([H])C([H])=C1[H] 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- LLBZPESJRQGYMB-UHFFFAOYSA-N 4-one Natural products O1C(C(=O)CC)CC(C)C11C2(C)CCC(C3(C)C(C(C)(CO)C(OC4C(C(O)C(O)C(COC5C(C(O)C(O)CO5)OC5C(C(OC6C(C(O)C(O)C(CO)O6)O)C(O)C(CO)O5)OC5C(C(O)C(O)C(C)O5)O)O4)O)CC3)CC3)=C3C2(C)CC1 LLBZPESJRQGYMB-UHFFFAOYSA-N 0.000 description 1
- GWCPOASUTWKBIK-UHFFFAOYSA-N 7-bromo-1-(2-methylpropyl)-2-sulfanylidene-5h-pyrrolo[3,2-d]pyrimidin-4-one Chemical compound O=C1NC(=S)N(CC(C)C)C2=C1NC=C2Br GWCPOASUTWKBIK-UHFFFAOYSA-N 0.000 description 1
- OPZUKVWMKYAGPB-UHFFFAOYSA-N 7-methyl-3-(2-methylpropyl)-2-sulfanylidenepurin-6-one Chemical compound O=C1NC(=S)N(CC(C)C)C2=C1N(C)C=N2 OPZUKVWMKYAGPB-UHFFFAOYSA-N 0.000 description 1
- WLSCEYDWCKJQSG-UHFFFAOYSA-N 8-methyl-3-(2-methylpropyl)-2-sulfanylidene-7h-purin-6-one Chemical compound O=C1NC(=S)N(CC(C)C)C2=C1NC(C)=N2 WLSCEYDWCKJQSG-UHFFFAOYSA-N 0.000 description 1
- IXHYELJKKHRULV-UHFFFAOYSA-N 8-methyl-3-(2-methylpropyl)-6-sulfanylidene-7h-purin-2-one Chemical compound S=C1NC(=O)N(CC(C)C)C2=C1NC(C)=N2 IXHYELJKKHRULV-UHFFFAOYSA-N 0.000 description 1
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical group CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 1
- 108010005094 Advanced Glycation End Products Proteins 0.000 description 1
- 108010051842 Advanced Oxidation Protein Products Proteins 0.000 description 1
- USFZMSVCRYTOJT-UHFFFAOYSA-N Ammonium acetate Chemical compound N.CC(O)=O USFZMSVCRYTOJT-UHFFFAOYSA-N 0.000 description 1
- 239000005695 Ammonium acetate Substances 0.000 description 1
- 108020004491 Antisense DNA Proteins 0.000 description 1
- 108020005544 Antisense RNA Proteins 0.000 description 1
- 206010002961 Aplasia Diseases 0.000 description 1
- 206010003210 Arteriosclerosis Diseases 0.000 description 1
- 206010003225 Arteriospasm coronary Diseases 0.000 description 1
- XUKUURHRXDUEBC-KAYWLYCHSA-N Atorvastatin Chemical compound C=1C=CC=CC=1C1=C(C=2C=CC(F)=CC=2)N(CC[C@@H](O)C[C@@H](O)CC(O)=O)C(C(C)C)=C1C(=O)NC1=CC=CC=C1 XUKUURHRXDUEBC-KAYWLYCHSA-N 0.000 description 1
- XUKUURHRXDUEBC-UHFFFAOYSA-N Atorvastatin Natural products C=1C=CC=CC=1C1=C(C=2C=CC(F)=CC=2)N(CCC(O)CC(O)CC(O)=O)C(C(C)C)=C1C(=O)NC1=CC=CC=C1 XUKUURHRXDUEBC-UHFFFAOYSA-N 0.000 description 1
- 238000009020 BCA Protein Assay Kit Methods 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 102000049320 CD36 Human genes 0.000 description 1
- 108010045374 CD36 Antigens Proteins 0.000 description 1
- 102000000905 Cadherin Human genes 0.000 description 1
- 108050007957 Cadherin Proteins 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 1
- 108020004705 Codon Proteins 0.000 description 1
- 108010051219 Cre recombinase Proteins 0.000 description 1
- 102100025620 Cytochrome b-245 light chain Human genes 0.000 description 1
- 101710190086 Cytochrome b-245 light chain Proteins 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 241000991587 Enterovirus C Species 0.000 description 1
- 102000006396 Ephrin-B2 Human genes 0.000 description 1
- 108010044090 Ephrin-B2 Proteins 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 208000031448 Genomic Instability Diseases 0.000 description 1
- ZWZWYGMENQVNFU-UHFFFAOYSA-N Glycerophosphorylserin Natural products OC(=O)C(N)COP(O)(=O)OCC(O)CO ZWZWYGMENQVNFU-UHFFFAOYSA-N 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 206010062713 Haemorrhagic diathesis Diseases 0.000 description 1
- 241000713858 Harvey murine sarcoma virus Species 0.000 description 1
- 206010019280 Heart failures Diseases 0.000 description 1
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 1
- 208000036066 Hemophagocytic Lymphohistiocytosis Diseases 0.000 description 1
- 208000024659 Hemostatic disease Diseases 0.000 description 1
- 206010019842 Hepatomegaly Diseases 0.000 description 1
- 208000032672 Histiocytosis haematophagic Diseases 0.000 description 1
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 1
- 101001099460 Homo sapiens Myeloperoxidase Proteins 0.000 description 1
- 101000588302 Homo sapiens Nuclear factor erythroid 2-related factor 2 Proteins 0.000 description 1
- 101000997835 Homo sapiens Tyrosine-protein kinase JAK1 Proteins 0.000 description 1
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 1
- 102000004286 Hydroxymethylglutaryl CoA Reductases Human genes 0.000 description 1
- 108090000895 Hydroxymethylglutaryl CoA Reductases Proteins 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
- 102000042838 JAK family Human genes 0.000 description 1
- 108091082332 JAK family Proteins 0.000 description 1
- 108010019437 Janus Kinase 2 Proteins 0.000 description 1
- 206010072206 Janus kinase 2 mutation Diseases 0.000 description 1
- 102100020675 Krueppel-like factor 2 Human genes 0.000 description 1
- 101710186679 Kruppel-like factor 2 Proteins 0.000 description 1
- 238000012313 Kruskal-Wallis test Methods 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 1
- 208000037380 MINOCA Diseases 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 241000713869 Moloney murine leukemia virus Species 0.000 description 1
- PCZOHLXUXFIOCF-UHFFFAOYSA-N Monacolin X Natural products C12C(OC(=O)C(C)CC)CC(C)C=C2C=CC(C)C1CCC1CC(O)CC(=O)O1 PCZOHLXUXFIOCF-UHFFFAOYSA-N 0.000 description 1
- 102100031701 Nuclear factor erythroid 2-related factor 2 Human genes 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 101150117945 PDGFB gene Proteins 0.000 description 1
- 241001631646 Papillomaviridae Species 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- 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 1
- 229940122907 Phosphatase inhibitor Drugs 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 208000008601 Polycythemia Diseases 0.000 description 1
- 108010093965 Polymyxin B Proteins 0.000 description 1
- 241001505332 Polyomavirus sp. Species 0.000 description 1
- 201000009454 Portal vein thrombosis Diseases 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- TUZYXOIXSAXUGO-UHFFFAOYSA-N Pravastatin Natural products C1=CC(C)C(CCC(O)CC(O)CC(O)=O)C2C(OC(=O)C(C)CC)CC(O)C=C21 TUZYXOIXSAXUGO-UHFFFAOYSA-N 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 108010026552 Proteome Proteins 0.000 description 1
- 239000012083 RIPA buffer Substances 0.000 description 1
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 1
- 108091030071 RNAI Proteins 0.000 description 1
- 108010091086 Recombinases Proteins 0.000 description 1
- 102000018120 Recombinases Human genes 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 235000014548 Rubus moluccanus Nutrition 0.000 description 1
- 108091005623 S-nitrosylated proteins Proteins 0.000 description 1
- BQCADISMDOOEFD-UHFFFAOYSA-N Silver Chemical compound [Ag] BQCADISMDOOEFD-UHFFFAOYSA-N 0.000 description 1
- KEAYESYHFKHZAL-UHFFFAOYSA-N Sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 1
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 1
- 206010049418 Sudden Cardiac Death Diseases 0.000 description 1
- 208000001871 Tachycardia Diseases 0.000 description 1
- 241000053227 Themus Species 0.000 description 1
- ZMZDMBWJUHKJPS-UHFFFAOYSA-M Thiocyanate anion Chemical compound [S-]C#N ZMZDMBWJUHKJPS-UHFFFAOYSA-M 0.000 description 1
- 102100033438 Tyrosine-protein kinase JAK1 Human genes 0.000 description 1
- 102000008790 VE-cadherin Human genes 0.000 description 1
- 241000700618 Vaccinia virus Species 0.000 description 1
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 1
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 1
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 1
- 208000035868 Vascular inflammations Diseases 0.000 description 1
- 206010047139 Vasoconstriction Diseases 0.000 description 1
- 206010047141 Vasodilatation Diseases 0.000 description 1
- 206010047281 Ventricular arrhythmia Diseases 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 230000021736 acetylation Effects 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- YJVBLROMQZEFPA-UHFFFAOYSA-L acid red 26 Chemical compound [Na+].[Na+].CC1=CC(C)=CC=C1N=NC1=C(O)C(S([O-])(=O)=O)=CC2=CC(S([O-])(=O)=O)=CC=C12 YJVBLROMQZEFPA-UHFFFAOYSA-L 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 206010000891 acute myocardial infarction Diseases 0.000 description 1
- 239000003454 advanced oxidation protein product Substances 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 238000012152 algorithmic method Methods 0.000 description 1
- 230000000172 allergic effect Effects 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 235000001014 amino acid Nutrition 0.000 description 1
- 235000019257 ammonium acetate Nutrition 0.000 description 1
- 229940043376 ammonium acetate Drugs 0.000 description 1
- 230000033115 angiogenesis Effects 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000692 anti-sense effect Effects 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 239000003816 antisense DNA Substances 0.000 description 1
- 230000001640 apoptogenic effect Effects 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 206010003119 arrhythmia Diseases 0.000 description 1
- 230000006793 arrhythmia Effects 0.000 description 1
- 208000011775 arteriosclerosis disease Diseases 0.000 description 1
- 208000028922 artery disease Diseases 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 238000003149 assay kit Methods 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 229960005370 atorvastatin Drugs 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 229920002988 biodegradable polymer Polymers 0.000 description 1
- 239000004621 biodegradable polymer Substances 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 210000003995 blood forming stem cell Anatomy 0.000 description 1
- 230000013926 blood microparticle formation Effects 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 238000010322 bone marrow transplantation Methods 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 238000009395 breeding Methods 0.000 description 1
- 230000001488 breeding effect Effects 0.000 description 1
- 238000010804 cDNA synthesis Methods 0.000 description 1
- 108010018828 cadherin 5 Proteins 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 210000000748 cardiovascular system Anatomy 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 230000011748 cell maturation Effects 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 230000005754 cellular signaling Effects 0.000 description 1
- 230000007213 cerebrovascular event Effects 0.000 description 1
- 229960005110 cerivastatin Drugs 0.000 description 1
- SEERZIQQUAZTOL-ANMDKAQQSA-N cerivastatin Chemical compound COCC1=C(C(C)C)N=C(C(C)C)C(\C=C\[C@@H](O)C[C@@H](O)CC(O)=O)=C1C1=CC=C(F)C=C1 SEERZIQQUAZTOL-ANMDKAQQSA-N 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 150000005829 chemical entities Chemical class 0.000 description 1
- 235000012000 cholesterol Nutrition 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 210000003040 circulating cell Anatomy 0.000 description 1
- 230000015271 coagulation Effects 0.000 description 1
- 238000005345 coagulation Methods 0.000 description 1
- 230000001332 colony forming effect Effects 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 239000003184 complementary RNA Substances 0.000 description 1
- 238000004624 confocal microscopy Methods 0.000 description 1
- 239000000356 contaminant Substances 0.000 description 1
- 108010009442 cytochrome b245 Proteins 0.000 description 1
- 230000009089 cytolysis Effects 0.000 description 1
- 230000002435 cytoreductive effect Effects 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 230000007547 defect Effects 0.000 description 1
- 230000006735 deficit Effects 0.000 description 1
- 229960003964 deoxycholic acid Drugs 0.000 description 1
- 230000002074 deregulated effect Effects 0.000 description 1
- 230000003831 deregulation Effects 0.000 description 1
- 238000011033 desalting Methods 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 238000010586 diagram Methods 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 230000009699 differential effect Effects 0.000 description 1
- 230000005750 disease progression Effects 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
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 230000037437 driver mutation Effects 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 241001493065 dsRNA viruses Species 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 238000001378 electrochemiluminescence detection Methods 0.000 description 1
- 230000000925 erythroid effect Effects 0.000 description 1
- 230000000763 evoking effect Effects 0.000 description 1
- 230000005284 excitation Effects 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 238000013401 experimental design Methods 0.000 description 1
- 210000001723 extracellular space Anatomy 0.000 description 1
- 230000020764 fibrinolysis Effects 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 238000000684 flow cytometry Methods 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- 229960003765 fluvastatin Drugs 0.000 description 1
- 238000013467 fragmentation Methods 0.000 description 1
- 238000006062 fragmentation reaction Methods 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 238000002546 full scan Methods 0.000 description 1
- 239000007789 gas Substances 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 210000003714 granulocyte Anatomy 0.000 description 1
- 150000004820 halides Chemical class 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 108010036302 hemoglobin AS Proteins 0.000 description 1
- 208000014752 hemophagocytic syndrome Diseases 0.000 description 1
- 208000031169 hemorrhagic disease Diseases 0.000 description 1
- 230000023597 hemostasis Effects 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- ZFGMDIBRIDKWMY-PASTXAENSA-N heparin Chemical compound CC(O)=N[C@@H]1[C@@H](O)[C@H](O)[C@@H](COS(O)(=O)=O)O[C@@H]1O[C@@H]1[C@@H](C(O)=O)O[C@@H](O[C@H]2[C@@H]([C@@H](OS(O)(=O)=O)[C@@H](O[C@@H]3[C@@H](OC(O)[C@H](OS(O)(=O)=O)[C@H]3O)C(O)=O)O[C@@H]2O)CS(O)(=O)=O)[C@H](O)[C@H]1O ZFGMDIBRIDKWMY-PASTXAENSA-N 0.000 description 1
- 229960001008 heparin sodium Drugs 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- 102000051251 human MPO Human genes 0.000 description 1
- ZMZDMBWJUHKJPS-UHFFFAOYSA-N hydrogen thiocyanate Natural products SC#N ZMZDMBWJUHKJPS-UHFFFAOYSA-N 0.000 description 1
- 230000007062 hydrolysis Effects 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- QWPPOHNGKGFGJK-UHFFFAOYSA-N hypochlorous acid Chemical compound ClO QWPPOHNGKGFGJK-UHFFFAOYSA-N 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 230000007574 infarction Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000007917 intracranial administration Methods 0.000 description 1
- 229960004359 iodixanol Drugs 0.000 description 1
- PGLTVOMIXTUURA-UHFFFAOYSA-N iodoacetamide Chemical compound NC(=O)CI PGLTVOMIXTUURA-UHFFFAOYSA-N 0.000 description 1
- PGHMRUGBZOYCAA-ADZNBVRBSA-N ionomycin Chemical compound O1[C@H](C[C@H](O)[C@H](C)[C@H](O)[C@H](C)/C=C/C[C@@H](C)C[C@@H](C)C(/O)=C/C(=O)[C@@H](C)C[C@@H](C)C[C@@H](CCC(O)=O)C)CC[C@@]1(C)[C@@H]1O[C@](C)([C@@H](C)O)CC1 PGHMRUGBZOYCAA-ADZNBVRBSA-N 0.000 description 1
- 238000003368 label free method Methods 0.000 description 1
- 238000012332 laboratory investigation Methods 0.000 description 1
- 210000002414 leg Anatomy 0.000 description 1
- 230000003902 lesion Effects 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 230000001665 lethal effect Effects 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 229960004844 lovastatin Drugs 0.000 description 1
- PCZOHLXUXFIOCF-BXMDZJJMSA-N lovastatin Chemical compound C([C@H]1[C@@H](C)C=CC2=C[C@H](C)C[C@@H]([C@H]12)OC(=O)[C@@H](C)CC)C[C@@H]1C[C@@H](O)CC(=O)O1 PCZOHLXUXFIOCF-BXMDZJJMSA-N 0.000 description 1
- QLJODMDSTUBWDW-UHFFFAOYSA-N lovastatin hydroxy acid Natural products C1=CC(C)C(CCC(O)CC(O)CC(O)=O)C2C(OC(=O)C(C)CC)CC(C)C=C21 QLJODMDSTUBWDW-UHFFFAOYSA-N 0.000 description 1
- 230000035168 lymphangiogenesis Effects 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 229910052943 magnesium sulfate Inorganic materials 0.000 description 1
- 238000002826 magnetic-activated cell sorting Methods 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 235000012054 meals Nutrition 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 229930182817 methionine Natural products 0.000 description 1
- 239000011325 microbead Substances 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 229910000402 monopotassium phosphate Inorganic materials 0.000 description 1
- 229910000403 monosodium phosphate Inorganic materials 0.000 description 1
- 235000019799 monosodium phosphate Nutrition 0.000 description 1
- FEBLFNYEUJEOLD-UHFFFAOYSA-N n-[1-(6-oxo-2-sulfanylidene-7h-purin-3-yl)propan-2-yl]-1h-imidazole-2-carboxamide Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CC(C)NC(=O)C1=NC=CN1 FEBLFNYEUJEOLD-UHFFFAOYSA-N 0.000 description 1
- SMBFHTPOCAIXGA-UHFFFAOYSA-N n-[1-(6-oxo-2-sulfanylidene-7h-purin-3-yl)propan-2-yl]pyridine-2-carboxamide Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CC(C)NC(=O)C1=CC=CC=N1 SMBFHTPOCAIXGA-UHFFFAOYSA-N 0.000 description 1
- GSLCARCSLQLGFD-UHFFFAOYSA-N n-[1-(6-oxo-2-sulfanylidene-7h-purin-3-yl)propan-2-yl]pyridine-3-carboxamide Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CC(C)NC(=O)C1=CC=CN=C1 GSLCARCSLQLGFD-UHFFFAOYSA-N 0.000 description 1
- KCYZTORODXQMMU-UHFFFAOYSA-N n-[1-(6-oxo-2-sulfanylidene-7h-purin-3-yl)propan-2-yl]pyrimidine-2-carboxamide Chemical compound C1=2N=CNC=2C(=O)NC(=S)N1CC(C)NC(=O)C1=NC=CC=N1 KCYZTORODXQMMU-UHFFFAOYSA-N 0.000 description 1
- CVVVWYUXSNNDLD-UHFFFAOYSA-N n-[1-(6-oxo-2-sulfanylidene-7h-purin-3-yl)propan-2-yl]quinoline-2-carboxamide Chemical compound C1=CC=CC2=NC(C(=O)NC(CN3C(NC(=O)C=4NC=NC=43)=S)C)=CC=C21 CVVVWYUXSNNDLD-UHFFFAOYSA-N 0.000 description 1
- 239000002840 nitric oxide donor Substances 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 210000004940 nucleus Anatomy 0.000 description 1
- 238000003305 oral gavage Methods 0.000 description 1
- 238000012261 overproduction Methods 0.000 description 1
- 239000007800 oxidant agent Substances 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- 239000013610 patient sample Substances 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 239000002831 pharmacologic agent Substances 0.000 description 1
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 1
- COLNVLDHVKWLRT-QMMMGPOBSA-N phenylalanine group Chemical class N[C@@H](CC1=CC=CC=C1)C(=O)O COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 1
- 229960003733 phenylephrine hydrochloride Drugs 0.000 description 1
- OCYSGIYOVXAGKQ-FVGYRXGTSA-N phenylephrine hydrochloride Chemical compound [H+].[Cl-].CNC[C@H](O)C1=CC=CC(O)=C1 OCYSGIYOVXAGKQ-FVGYRXGTSA-N 0.000 description 1
- 150000004713 phosphodiesters Chemical class 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- 239000002504 physiological saline solution Substances 0.000 description 1
- 229940096701 plain lipid modifying drug hmg coa reductase inhibitors Drugs 0.000 description 1
- 239000013612 plasmid Substances 0.000 description 1
- 229920000410 poly[9,9-bis((6'-N,N,N-trimethylammonium)hexyl)fluorenylene phenylene dibromide] polymer Polymers 0.000 description 1
- 229920000024 polymyxin B Polymers 0.000 description 1
- 229960005266 polymyxin b Drugs 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 229960002965 pravastatin Drugs 0.000 description 1
- TUZYXOIXSAXUGO-PZAWKZKUSA-N pravastatin Chemical compound C1=C[C@H](C)[C@H](CC[C@@H](O)C[C@@H](O)CC(O)=O)[C@H]2[C@@H](OC(=O)[C@@H](C)CC)C[C@H](O)C=C21 TUZYXOIXSAXUGO-PZAWKZKUSA-N 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 239000003805 procoagulant Substances 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 238000000575 proteomic method Methods 0.000 description 1
- 125000002577 pseudohalo group Chemical group 0.000 description 1
- UYLWKSJTHLRFBX-UHFFFAOYSA-N purin-6-one Chemical compound O=C1N=CN=C2N=CN=C12 UYLWKSJTHLRFBX-UHFFFAOYSA-N 0.000 description 1
- 239000002096 quantum dot Substances 0.000 description 1
- 150000003254 radicals Chemical class 0.000 description 1
- 238000004725 rapid separation liquid chromatography Methods 0.000 description 1
- 208000012284 reactive thrombocytosis Diseases 0.000 description 1
- 239000001044 red dye Substances 0.000 description 1
- 230000011514 reflex Effects 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 238000007634 remodeling Methods 0.000 description 1
- 230000010410 reperfusion Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000011506 response to oxidative stress Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 238000012502 risk assessment Methods 0.000 description 1
- 229960000672 rosuvastatin Drugs 0.000 description 1
- BPRHUIZQVSMCRT-VEUZHWNKSA-N rosuvastatin Chemical compound CC(C)C1=NC(N(C)S(C)(=O)=O)=NC(C=2C=CC(F)=CC=2)=C1\C=C\[C@@H](O)C[C@@H](O)CC(O)=O BPRHUIZQVSMCRT-VEUZHWNKSA-N 0.000 description 1
- 230000002000 scavenging effect Effects 0.000 description 1
- 238000012216 screening Methods 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
- 229910052709 silver Inorganic materials 0.000 description 1
- 239000004332 silver Substances 0.000 description 1
- 238000001542 size-exclusion chromatography Methods 0.000 description 1
- 210000000329 smooth muscle myocyte Anatomy 0.000 description 1
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 1
- FHHPUSMSKHSNKW-SMOYURAASA-M sodium deoxycholate Chemical compound [Na+].C([C@H]1CC2)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC([O-])=O)C)[C@@]2(C)[C@@H](O)C1 FHHPUSMSKHSNKW-SMOYURAASA-M 0.000 description 1
- ZNJHFNUEQDVFCJ-UHFFFAOYSA-M sodium;2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid;hydroxide Chemical compound [OH-].[Na+].OCCN1CCN(CCS(O)(=O)=O)CC1 ZNJHFNUEQDVFCJ-UHFFFAOYSA-M 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 230000002966 stenotic effect Effects 0.000 description 1
- 230000001954 sterilising effect Effects 0.000 description 1
- 238000004659 sterilization and disinfection Methods 0.000 description 1
- 239000011550 stock solution Substances 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- CCEKAJIANROZEO-UHFFFAOYSA-N sulfluramid Chemical group CCNS(=O)(=O)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)F CCEKAJIANROZEO-UHFFFAOYSA-N 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 208000011580 syndromic disease Diseases 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000002123 temporal effect Effects 0.000 description 1
- 201000005665 thrombophilia Diseases 0.000 description 1
- 210000002303 tibia Anatomy 0.000 description 1
- 125000002088 tosyl group Chemical group [H]C1=C([H])C(=C([H])C([H])=C1C([H])([H])[H])S(*)(=O)=O 0.000 description 1
- 238000012085 transcriptional profiling Methods 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- HRXKRNGNAMMEHJ-UHFFFAOYSA-K trisodium citrate Chemical compound [Na+].[Na+].[Na+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O HRXKRNGNAMMEHJ-UHFFFAOYSA-K 0.000 description 1
- 235000019263 trisodium citrate Nutrition 0.000 description 1
- 229940038773 trisodium citrate Drugs 0.000 description 1
- GPRLSGONYQIRFK-MNYXATJNSA-N triton Chemical compound [3H+] GPRLSGONYQIRFK-MNYXATJNSA-N 0.000 description 1
- 210000003606 umbilical vein Anatomy 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 238000001291 vacuum drying Methods 0.000 description 1
- 238000010200 validation analysis Methods 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- LSGOVYNHVSXFFJ-UHFFFAOYSA-N vanadate(3-) Chemical class [O-][V]([O-])([O-])=O LSGOVYNHVSXFFJ-UHFFFAOYSA-N 0.000 description 1
- 210000003556 vascular endothelial cell Anatomy 0.000 description 1
- 210000004509 vascular smooth muscle cell Anatomy 0.000 description 1
- 239000002550 vasoactive agent Substances 0.000 description 1
- 230000025033 vasoconstriction Effects 0.000 description 1
- 230000024883 vasodilation Effects 0.000 description 1
- 229940124549 vasodilator Drugs 0.000 description 1
- 239000003071 vasodilator agent Substances 0.000 description 1
- 230000002455 vasospastic effect Effects 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
- 238000012795 verification Methods 0.000 description 1
- 239000013603 viral vector Substances 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 230000002747 voluntary effect Effects 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 238000007482 whole exome sequencing Methods 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
- A61K31/52—Purines, e.g. adenine
- A61K31/522—Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6876—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
- C12Q1/6883—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/569—Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
- G01N33/56966—Animal cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q2600/00—Oligonucleotides characterized by their use
- C12Q2600/156—Polymorphic or mutational markers
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2800/00—Detection or diagnosis of diseases
- G01N2800/32—Cardiovascular disorders
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2800/00—Detection or diagnosis of diseases
- G01N2800/50—Determining the risk of developing a disease
Definitions
- the present invention is in the field of medicine.
- Bcr/Abl-negative myeloproliferative neoplasms are clonal hematopoietic stem cell disorders characterized by the proliferation of particular hematopoietic lineages without blockage in cell maturation. They include polycythemia vera, essential thrombocythemia, and primary myelofibrosis (1).
- JAK2 is the most common MPN driver gene. JAK2 V617F is a gain of function mutation leading to growth factors hypersensitivity, detected in around 70% of MPNs (95% in polycythemia vera and 50% to 60% in essential thrombocythemia and pre-primary myelofibrosis / primary myelofibrosis) (1).
- JAK2 V617F appears in pluripotent hematopoietic progenitor cells and is present in all erythroid and myeloid lineages (1).
- JAK2 V617F in endothelial cells in the liver and the spleen of patients with splanchnic vein thrombosis (2, 3) and in circulating endothelial progenitor cells (4 6).
- Cardiovascular diseases reveal MPNs in about 30% of the patients and are the first cause of morbidity and mortality in these patients (7).
- Arterial events represent 60-70% of these cardio-vascular events (7).
- myocardial infarction without significant coronary stenosis by angiography was observed in 21% of patients with MNP (8) versus only 3% in a similar population without MPN (9).
- This observation prompted the European society of cardiology to recommend searching for MPNs in case of myocardial infarction without obstructive coronary artery disease (10).
- the mechanism underlying this link between myocardial infarction without obstructive coronary artery disease and MPNs is unknown, but vasoactive phenomenon (local intense vasoconstriction) can be suspected (11, 12).
- the present invention relates to the use of myeloperoxidase inhibitors for the treatment of cardiovascular diseases in patients suffering from myeloproliferative neoplasms.
- the inventors identified a high expression of myeloperoxidase in microvesicles derived from JAK2 V617F erythrocytes that could account for this effect. To assess the role of myeloperoxidase in this effect, the inventors then directly inhibited myeloperoxidase in microvesicles derived from Jak2 V6I7F erythrocytes and observed that it completely reversed the increase in endothelial oxidative stress induced by microvesicles derived from Jak2 V6I7F .
- JAK2 V6I7F MPN induce a potent increase in arterial contraction with increased endothelial oxidative stress, mediated by erythrocytes microvesicles and that myeloperoxidase (MPO) inhibitors would be suitable for preventing cardiovascular diseases in patient suffering from MPN.
- MPO myeloperoxidase
- the first object of the present invention relates to a method of preventing the occurrence of cardiovascular event in a patient suffering from a myeloproliferative neoplasm comprising administering to the patient a therapeutically effective amount of a myeloperoxidase inhibitor.
- MPN myeloproliferative neoplasm
- MPNs typically include polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF).
- PV polycythemia vera
- ET essential thrombocythemia
- PMF primary myelofibrosis
- IWG-MRT myeloproliferative neoplasms research and treatment
- the patient harbours one mutation in JAK2.
- JAK2 has its general meaning in the art and refers to the Janus Kinase 2 protein.
- the amino acid sequence of human JAK2 is well known in the art. Human JAK2 sequences are, for example, represented in the NCBI database (www.ncbi.orgwww.ncbi.nlm.nih.gov/), for example, under accession number NP 004963.
- Typical MPD associated mutation is the Jak2 V6I7F mutation which refers to the point mutation (1849 G for T) in exon 14, which causes the substitution of phenylalanine for valine at codon 617 in the JAK homology JH2 domain.
- JAK2 mutations include exon 12 mutations which can be substitutions, deletions, insertions and duplications, and all occur within a 44 nucleotides region in the JAK2 gene which encompasses amino acids 533-547 at the protein level.
- the most commonly reported mutations are small in-frame deletions of 3-12 nucleotides with a six nucleotides deletion being the most frequent. Complex mutations are present in one-third of cases with some mutations occurring outside this hotspot region.
- the N542-E543del is the most common mutation (23-30%), the K537L, E543-D544del and F537-K39delinsL represent 10-14%, and R541-E543delinsK comprise less than 10% of these mutations.
- JAK2 exon 12 mutations are located in a region close to the pseudo-kinase domain which acts as a linker between this domain and the Src homology 2 domain of JAK2.
- cardiovascular event refers to any disorder of the cardiovascular system including preferably any acute cardiovascular event.
- Acute cardiovascular events are, preferably, stable angina pectoris (SAP) or acute coronary syndrome (ACS).
- ACS patients can show unstable angina pectoris (UAP) or myocardial infarction (MI).
- MI can be an ST-elevation MI (STEMI) or a non-ST-elevation MI (NSTEMI).
- NSTE-ACS as used herein encompasses UAP and NSTEMI.
- the occurring of an MI can be followed by a left ventricular dysfunction (LVD), development of heart failure or even mortality.
- Further preferred cardiovascular events encompass cardiac brady- or tachyarrhythmias including sudden cardiac death and stroke (cerebrovascular events or accidents).
- mortality can also refer to the death rate or the ratio of number of deaths to a given population of subjects.
- MPO Myeloperoxidase
- MPO has its general meaning in the art and refers to a heme-containing enzyme.
- the enzyme uses hydrogen peroxide to oxidize chloride to hypochlorous acid.
- Other halides and pseudohalides like thiocyanate are also physiological substrates to MPO.
- MPO inhibitor refers to any compound natural or not which is capable of inhibiting the activity of MPO, in particular MPO kinase activity.
- MPO inhibitors are well known in the art.
- the term encompasses any MPO inhibitor that is currently known in the art or that will be identified in the future, and includes any chemical entity that, upon administration to a patient, results in inhibition or down-regulation of a biological activity associated with activation of the MPO.
- the term also encompasses inhibitor of expression.
- the MPO inhibition of the compounds may be determined using various methods well known in the art.
- Examples of compounds that can be used as MPO-inhibitors are compounds described in WO 2006/062465, WO 2006/062465, WO 2003/089430, WO 2003/089430, or WO 2003/089430.
- the MPO inhibitor of the present invention is selected from the group consisting of:
- the MPO inhibitor is AZD5904 which has the formula of:
- the MPO inhibitor is an inhibitor of MPO expression.
- An “inhibitor of expression” refers to a natural or synthetic compound that has a biological effect to inhibit the expression of a gene.
- said inhibitor of gene expression is a siRNA, an antisense oligonucleotide or a ribozyme.
- anti- sense oligonucleotides including anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of MPO mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of MPO, and thus activity, in a cell.
- antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence encoding MPO 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).
- Small inhibitory RNAs siRNAs
- siRNAs can also function as inhibitors of expression for use in the present invention.
- MPO gene expression can be reduced by contacting a subject or cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that MPO gene expression is specifically inhibited (i.e. RNA interference or RNAi).
- dsRNA small double stranded RNA
- RNAi RNA interference or RNAi
- Antisense oligonucleotides, siRNAs, shRNAs and ribozymes 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 cells expressing MPO.
- 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; SV40-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
- SV40-type viruses polyoma viruses
- Epstein-Barr viruses Epstein-Barr viruses
- papilloma viruses herpes virus
- vaccinia virus
- a “therapeutically effective amount” of the inhibitor as above described is meant a sufficient amount to provide a therapeutic effect. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
- the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed, the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide employed; and like factors well known in the medical arts.
- the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day.
- the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject to be treated.
- a medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from 1 mg to about 100 mg of the active ingredient.
- An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
- the inhibitor of the present invention is combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form pharmaceutical compositions.
- pharmaceutically acceptable excipients such as biodegradable polymers
- 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.
- saline solutions monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts
- 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.
- Sterile injectable solutions are prepared by incorporating the inhibitor at 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.
- 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.
- a further object of the present invention relates to a method of determining whether a patient suffering from a myeloproliferative neoplasm is at risk of having a cardiovascular event comprising the steps of determining the level of microvesicles derived from Jak2 V6!7F erythrocytes in a blood sample obtained from the patient wherein the level correlates with the risk of having a cardiovascular disease.
- the term“risk” relates to the probability that an event will occur over a specific time period, as in the conversion to a cardiovascular event, and can mean a subject's "absolute" risk or "relative” risk.
- Absolute risk can be measured with reference to either actual observation post-measurement for the relevant time cohort, or with reference to index values developed from statistically valid historical cohorts that have been followed for the relevant time period.
- Relative risk refers to the ratio of absolute risks of a subject compared either to the absolute risks of low risk cohorts or an average population risk, which can vary by how clinical risk factors are assessed. Odds ratios, the proportion of positive events to negative events for a given test result, are also commonly used (odds are according to the formula p/(l-p) where p is the probability of event and (1- p) is the probability of no event) to no- conversion.
- Alternative continuous measures which may be assessed in the context of the present invention include time to a cardiovascular event conversion and therapeutic a cardiovascular event conversion risk reduction ratios.
- determining whether a patient is at risk of having a cardiovascular event means that the patient to be analyzed by the method of the present invention is allocated either into the group of patients of a population having an elevated risk, or into a group having a reduced risk of having a cardiovascular event.
- An elevated risk as referred to in accordance with the present invention, preferably, means that the risk of developing a cardiovascular event within a predetermined predictive window is elevated significantly (i.e. increased significantly) for a patient with respect to the average risk for a cardiovascular event or cardiac mortality in a population of patients.
- a reduced risk as referred to in accordance with the present invention preferably, means that the risk of developing a cardiovascular event within a predetermined predictive window is reduced significantly for a patient with respect to the average risk for a cardiovascular event or cardiac mortality in a population of patients.
- a significant increase or reduction of a risk is an increase or reduction or a risk of a size which is considered to be significant for prognosis, particularly said increase or reduction is considered statistically significant.
- the terms "significant” and "statistically significant" are known by the person skilled in the art. Thus, whether an increase or reduction of a risk is significant or statistically significant can be determined without further ado by the person skilled in the art using various well known statistic evaluation tools.
- blood sample means a whole blood, serum, or plasma sample obtained from the patient.
- the blood sample is a plasma sample.
- a plasma sample may be obtained using methods well known in the art. For example, blood may be drawn from the patient following standard venipuncture procedure on tri-sodium citrate buffer. Plasma may then be obtained from the blood sample following standard procedures including but not limited to, centrifuging the blood sample at about 2500*g for about 15 minutes (room temperature), followed by pipetting of the plasma layer. Platelet-free plasma (PFP) will be obtained following a second centrifugation at about 2500*g for 15 min. Analyses can be performed directly on this PFP.
- PFP Platelet-free plasma
- microvesicles may be more specifically isolated by further centrifuging the PFP at about 15,000 to about 25,000*g at 4°C.
- Different buffers may be considered appropriate for resuspending the pelleted cellular debris which contains the MPs.
- buffers include reagent grade (distilled or deionized) water and phosphate buffered saline (PBS) pH 7.4 or NaCl 0.9%.
- PBS buffer Sheath fluid
- microvesicle or“MV” or“extracellular vesicles” has its general meaning in the art and denotes a plasma membrane vesicle shed from an apoptotic or activated cell.
- the size of microvesicles / extracellular vesicles ranges from 0.1 pm to 1 pm in diameter.
- the surface markers of microvesicles are the same as the cells from they originated.
- Standard methods for determining the level of microvesicles in a blood sample are well known in the art and typically involve the methods described in the EXAMPLE.
- the level of the microvesicles derived from Jak2 V617F erythrocytes is compared to a predetermined reference value.
- the predetermined reference value is a threshold value or a cut-off value.
- a "threshold value” or “cut off value” can be determined experimentally, empirically, or theoretically.
- a threshold value can also be arbitrarily selected based upon the existing experimental and/or clinical conditions, as would be recognized by a person of ordinary skilled in the art. For example, retrospective measurement of expression levels in properly banked historical patient samples may be used in establishing the predetermined reference value.
- the threshold value has to be determined in order to obtain the optimal sensitivity and specificity according to the function of the test and the benefit/risk balance (clinical consequences of false positive and false negative).
- the optimal sensitivity and specificity can be determined using a Receiver Operating Characteristic (ROC) curve based on experimental data. For example, after quantifying the expression level in a group of reference, one can use algorithmic analysis for the statistic treatment of the determined levels in samples to be tested, and thus obtain a classification standard having significance for sample classification.
- ROC curve Receiver Operator Characteristic Curve, which is also known as receiver operation characteristic curve. It is mainly used for clinical biochemical diagnostic tests.
- ROC curve is a comprehensive indicator that reflects the continuous variables of true positive rate (sensitivity) and false positive rate (1-specificity). It reveals the relationship between sensitivity and specificity with the image composition method.
- a series of different cut-off values are set as continuous variables to calculate a series of sensitivity and specificity values. Then sensitivity is used as the vertical coordinate and specificity is used as the horizontal coordinate to draw a curve. The higher the area under the curve (AUC), the higher the accuracy of diagnosis.
- AUC area under the curve
- the point closest to the far upper left of the coordinate diagram is a critical point having both high sensitivity and high specificity values.
- the AUC value of the ROC curve is between 1.0 and 0.5.
- AUC>0.5 the diagnostic result gets better and better as AUC approaches 1.
- AUC is between 0.5 and 0.7, the accuracy is low.
- AUC is between 0.7 and 0.9, the accuracy is moderate.
- AUC is higher than 0.9, the accuracy is quite high.
- This algorithmic method is preferably done with a computer.
- Existing software or systems in the art may be used for the drawing of the ROC curve, such as: MedCalc 9.2.0.1 medical statistical software, SPSS 9.0, ROCPOWER.SAS, DESIGNROC.FOR, MULTIREADER POWERSAS, CREATE-ROC.SAS, GB STAT VIO.O (Dynamic Microsystems, Inc. Silver Spring, Md., USA), etc.
- the predetermined reference value was established in a population of patients who did not have a cardiovascular event when blood was drawn. Accordingly when the level of microvesicles is higher than the predetermined reference value, it is concluded that the patient is at risk of having a cardiovascular event. On contrary, when the level of microvesicles is lower than the predetermined reference value, then is it concluded that the patient is not at risk of having a cardiovascular event.
- high statistical significance values are obtained for a range of successive arbitrary quantification values, and not only for a single arbitrary quantification value.
- a range of values is provided instead of using a definite predetermined reference value. Therefore, a minimal statistical significance value (minimal threshold of significance, e.g. maximal threshold P value) is arbitrarily set and a range of a plurality of arbitrary quantification values for which the statistical significance value calculated at step g) is higher (more significant, e.g. lower P value) are retained, so that a range of quantification values is provided.
- This range of quantification values includes a "cut-off 1 value as described above.
- a cut-off 1 value the outcome can be determined by comparing the expression level with the range of values which are identified.
- a cut-off value thus consists of a range of quantification values, e.g. centered on the quantification value for which the highest statistical significance value is found (e.g. generally the minimum p value which is found). For example, on a hypothetical scale of 1 to 10, if the ideal cut-off value (the value with the highest statistical significance) is 5, a suitable (exemplary) range may be from 4-6.
- a patient may be assessed by comparing values obtained by determining the level of microvesicles, where values greater than 5 reveal that the patient is at risk of having a cardiovascular event and values less than 5 reveal that the patient is not at risk of having a cardiovascular event.
- a patient may be assessed by comparing values obtained by measuring the level of microvesicles and comparing the values on a scale, where values above the range of 4-6 indicate that the patient is at risk of having a cardiovascular event and values below the range of 4-6 indicate that the patient is not at risk of having a cardiovascular event, with values falling within the range of 4-6 indicating an intermediate risk.
- the result given by the method of the invention may be used as a guide in determining how frequently a cardiovascular event should be screened, in selecting a therapy or treatment regimen for the patient.
- a cardiovascular event For example, when the patient has been determined as having a high risk of a cardiovascular event, he can be eligible for a therapy with a MPO inhibitor as described herein, and/or with a statin.
- the term“Statin” has its general meaning in the art and refers to a class of drugs that are inhibitors of HMG-CoA reductase.
- statin include but are not limited to pravastatin, fluvastatin, atorvastatin, lovastatin, simvastatin, rosuvastatin, and cerivastatin.
- Statins may be in the form of a salt, hydrate, solvate, polymorph, or a co-crystal. Statins may also be in the form of a hydrate, solvate, polymorph, or a co-crystal of a salt. Statins may also be present in the free acid or acetone form.
- FIGURES are a diagrammatic representation of FIGURES.
- Quantitative data are expressed as median with interquartile range and cumulative dose-response curves are expressed as mean with standard error of the mean. Abbreviations: * p ⁇ 0.05, *** p ⁇ 0.001; ns, not significant. Cumulative dose response curves and electrocardiogram recording were compared using an analysis of variance for repeated measures and other data were compared using the Mann- Whitney U-test. All tests were 2 sided.
- Quantitative data are expressed as median with interquartile range and cumulative dose-response curves are expressed as mean with standard error of the mean.
- Cumulative dose-response curve to phenylephrine of aortas from WT mice injected with vehicle (n 5) or with epoietin (n 8) (I).
- Quantitative data are expressed as median with interquartile range and cumulative dose-response curves are expressed as mean with standard error of the mean.
- Jak2 V617F HC-EC mice F
- control mice Jak2WT
- Jak2 i r ' r/I Flex/WT VE-Cadherin-cre-ERT2 mice
- Jak2 V6I7F EC G
- ROS reactive oxygen species
- mice were littermate, appropriate, age, sex and genetic background, matched to account for any variation in data. Institutional animal care and use committee at INSERM (Descartes university, Paris, France) approved all animal experiments (CEEA- 17053).
- mice were on a C57BL/6 background.
- Mice carrying constitutive Jak2 l Y ' 171 mutation in endothelial and hematopoietic cells were obtained by crossing VE-cadherin-Cre transgenic mice provided by M. Souyri (13) with J 2 (/577 Flex/WT mice provided by M. Villeval (50).
- Mice carrying inducible Jak2 V617F mutation specifically in endothelial cells were obtained by crossing VE-Cadherin-cre-ERT2 transgenic mice provided by R. Adams (51) with Jak2 V6I7F Flex/WT mice provided by M. Villeval (50).
- the Flex (for Flip-Excision) strategy allows the expression of a mutated gene in adulthood, in a temporal and tissue-specific manner (52). It allows an efficient and reliable Cre-mediated genetic switch: the expression of a given gene is turned on by inversion, while expression of another one is simultaneously turned off by excision. In all experiments, male and female mice were used.
- mice were euthanized between the ages of 8 and 17 weeks.
- mice were injected intraperitoneally with tamoxifen (Sigma, T5648), 1 mg/mouse/day for 5 consecutive days over 2 consecutive weeks (10 mg in total per mouse) between the ages of 5 to 7 weeks. Experiments were performed between 4 to 6 weeks after the last tamoxifen injection. Both female and male were used for each experiment.
- mice were on a C57BL/6 background. Mice with the mTmG reporter provided by C. James (Inserm 1034) were crossed with VE-cadherin-Cre transgenic mice provided by M. Souyri or VE-Cadherin-cre-ERT2 transgenic mice provided by R. Adams (51).
- mice were injected intraperitoneally with tamoxifen (Sigma, T5648), 1 mg/mice/day for 5 consecutive days over 2 consecutive weeks (10 mg in total per mice) between the ages of 5 to 7 weeks, and experiments were performed 2 weeks after the last injection of tamoxifen.
- Aortas and femurs were harvested under isoflurane anaesthesia and fixed in 4% PFA. Aorta were mounted“en face” on glass slides, while femurs were cryosectioned. All tissues were imaged using a Leica SP5 confocal microscope (Leica) at 400 X magnification.
- Leica Leica SP5 confocal microscope
- aortas from adult mice were isolated after animal sacrifice under 2% isoflurane anaesthesia.
- the aortic rings were mounted immediately in organ chambers (Multi WireMyograph system, model 610 M; Danish Myo Technology, Aarhus, Denmark) filled with Krebs-Ringer solution (NaCl 118.3 mmol/L, KC1 4.7 mmol/L, MgS04 1.2 mmol/L, KH2P04 1.2 mmol/L, CaC12 1.25 mmol/L, NaHC03 25.0 mmol/L and glucose 5 0 mmol/L) gassed with a mixture of 02 95% and C02 5% (pH 7.4).
- Krebs-Ringer solution NaCl 118.3 mmol/L, KC1 4.7 mmol/L, MgS04 1.2 mmol/L, KH2P04 1.2 mmol/L, CaC12 1.25 mmol/L, NaHC03 25.0 mmol/L and
- aorta rings were preincubated for 45 min with L-NAME 10-4 mol/L (Cayman, 80210) prior to concentration-response curve to phenylephrine without washout.
- L-NAME 10-4 mol/L Liyman, 80210
- the endothelium was mechanically removed by inserting the tip of a pair of forceps within the lumen and by gently rubbing the ring back and forth on a piece of wet tissue.
- NAC N- Acetyl- Cysteine experiment
- NAC was added to the Krebs-Ringer solution at a final concentration of 20 mmol/L.
- the femoral artery was carefully exposed from adult mice under 2% isoflurane anaesthesia.
- Circulating microvesicles from patients or healthy control were isolated from platelet- free plasma obtained by successive centrifugations of venous blood, as reported previously (53). Briefly, citrated venous blood (15 mL) was centrifuged twice at 2500g for 15 minutes (at room temperature) to remove cells and cell debris and to obtain platelet-free plasma (PFP). A portion of this PFP was then aliquoted and stored at -80°C. The rest was centrifuged at 20500g for 2 hours (4°C). Supernatant of this 20500g centrifugation was then discarded and the resulting microvesicles pellet was resuspended in a minimal volume of supernatant, aliquoted and stored at -80°C. For each patient, concentrations of annexin V positive microvesicles were analysed in the PFP and the resuspended pellet of microvesicles.
- Circulating levels of annexin V+ microvesicles were determined on a Gallios flow cytometer (Beckman Coulter, Villepinte, France) using a technique previously described in detail (49, 53).
- Plasma samples were collected from the inferior vena cava of Jak2 V617F HC-EC mice or littermate controls using a 25 G x G needle in a 1 mL syringe pre-coated with 3.8% sodium citrate.
- PFPs were generated as described above for patients and used to measure plasma annexin V positive microvesicles in mice.
- the pelleted cells obtained following the first 2500g centrifugation were resuspended in PBS to a final volume of 5 mL for control mice and 10 mL for Jak2 V6I7F HC-EC mice.
- PBMC, PMNC and erythrocytes were separated using a double percoll gradient (63% and 72% for control mice and 63% and 66% for Jak2 V l7! HC-EC mice) using a 700g centrifugation for 25 min, without brake.
- the slight differences between the protocols used for control and Jak2 V617F HC-EC mice are the results of the preliminary experiments we did to obtain pure isolation of each cell type.
- Cells were subsequently washed with PBS, then incubated with 5 gmol/L ionomycin TBS for 30min at 37°C to induce microvesicles generation. 5 mmol/L EDTA was then added to chelate free calcium.
- Microvesicles were isolated, as described above using a 20500g centrifugation during 45min. Concentrations of annexin V positive microvesicles (as described above) were analysed in the PFP and the 20500g microvesicles pellet for each mouse.
- Residual contaminating erythrocytes were removed by magnetic sorting. Briefly, the cell suspension was labelled with Anti-Ter-119 MicroBeads (Miltenyi Biotec ref 130-049-901) and erythrocytes (Ter- 119+) were negatively sorted using a MACS® Separator. The remaining cells (platelets) were subsequently washed with PBS and exposed to 5 pmol/L ionomycin in TBS for 30 minutes at 37°C. 5 mM EDTA was then added to chelate free calcium. Finally, cells were discarded by centrifugation at 15000g for 1 minute, the supernatant was collected and microvesicles isolated, as previously described.
- thoracic aortas from adult C57BL/6 mice (8 to 10 weeks old) were isolated after sacrifice under isoflurane anaesthesia.
- Mouse aortic rings were incubated for 24 hrs; 37°C in a 5% C02 incubator, with filtered DMEM supplemented with antibiotics (100 IU/mL streptomycin, 100 IU/mL penicillin (Gibco, Invitrogen, Paisley, Scotland), and 10 pg/mL polymyxin B (Sigma, St Louis, MO) in the presence of microvesicles.
- antibiotics 100 IU/mL streptomycin, 100 IU/mL penicillin (Gibco, Invitrogen, Paisley, Scotland)
- 10 pg/mL polymyxin B (Sigma, St Louis, MO)
- mice were injected intravenously (retro-orbital injection) with microvesicles (100 pL final volume with 2 pL heparin sodium (5000 IU/mL)). Experiments were performed 2 hours after injection.
- Microvesicles from patients and healthy controls were incubated at their respective individual plasma concentration (Annexin-V positive microvesicles).
- Microvesicles generated from mice were incubated or injected at the same final concentration for Juki 1 6171 HC-EC mice and control mice, namely 7000 Annexin V positive microvesicles / pL for erythrocyte and platelet-derived microvesicles and 700 Annexin V positive microvesicles / pL for PBMC and PMNC-derived microvesicles.
- mice have concentrations of Annexin V positive microvesicles between 1000 and 10000 / pL, and because PBMC and PMNC-derived microvesicles are consistently found less abundant in the blood than erythrocyte and platelet-derived microvesicles (27, 32).
- mice We subjected 6 to 8 weeks old C57B1/6J mice to medullar aplasia following 9.5 gray lethal total body irradiation.
- mice We repopulated the mice with an intravenous injection of bone marrow cells isolated from femurs and tibias of age matched Jak2 V617F HC-EC and of littermate control mice. Medullar reconstitution was allowed for 8 weeks before experiments.
- Hydroxyurea (Sigma, H8627), or the same volume of vehicle (NaCl 0.9%), was administrated for 10 consecutive days (100 mg/kg/day BID) by intra-peritoneal injections.
- Ruxolitinib (Novartis) was administered for 21 consecutive days (30 mg/kg 2 times per day) by oral gavage (54). Ruxolitinib was prepared from 15-mg commercial tablets in PEG300/5% dextrose mixed at a 1 :3 ratio, as previously reported (55). Control mice were administered the same volume of vehicle (PEG300/dextrose 5%).
- Simvastatin (Sigma S6196) was administered for 14 days (20 mg/kg/day, once a day) by intra-peritoneal injections. Activation by hydrolysis was first achieved by dissolving 50 mg in 1 mL of pure ethanol and adding 0.813 ml of 1 mol/L NaOH. pH was adjusted to 7.2 by adding small quantities of 1 mol/L HC1 and dilution was then performed in PBS (56). Control mice were injected with the same volume of vehicle. Human recombinant Epoietin alfa (5000 UEkg, diluted in 0.2% BSA in PBS) or vehicle (0.2% BSA in PBS) was administered to wild type mice every 2 days for 3 weeks by intraperitoneal injection, as previously described (57).
- N-Acetyl-Cysteine (commercial HIDONAC, Zambon) diluted in NaCl 0.9% or the same volume of vehicle (NaCl 0.9%), was administrated for 14 consecutive days (500 mg/kg/day) by intraperitoneal injections.
- CellROX® Deep Red Reagent is a fluorogenic probe designed to reliably measure reactive oxygen species inside living cells.
- the cell-permeable CellROX® Deep Red dye is nonfluorescent while outside of the cell and in a reduced state and, upon oxidation, exhibits excitation/emission maxima at 640/665 nm.
- samples were costained with DAPI (0.1 pg mL, Sigma) in order to identify cell nuclei.
- aortas were washed with PBS, mounted“en face” on glass slides and imaged using a bright field Zeiss Axio Imager Z1 (Zeiss) microscope. Images were acquired in the 2 hours following staining at 400 X magnification. CellROX® positive surface (in red) and the number of cells were quantified using Image J Software.
- HUVECs single donor, C-12200, lot 445Z011, PromoCell
- CELLROX® Fisher scientific, Cl 0422
- HUVECs were costained with DAPI (0.1 pg/mL, Sigma) in order to identify nuclei. Images were acquired using a Confocal microscope, Leica SP8 at 400 X magnification.
- Electrocardiograms were recorded from mice using the non-invasive ecgTUNNEL (Emka Technologies) with minimal filtering Electrocardiogram was continuously monitored for 3 min (baseline). Waveforms were recorded using Iox Software and heart rate and intervals were measured with ECG Auto from recording traces. Following baseline determination, the animals received a single administration of phenylephrine (bolus, 3mg/Kg) by intravenous route at the caudal vein and electrocardiogram were recording 3-5 minutes more.
- phenylephrine bolus, 3mg/Kg
- RNA gene allelic discrimination was performed by Taqman analysis with the ABI Prism GeneAmp 7500 Sequence Detection System (Applied Biosystem, Invitrogen) using as primers: TTTACAAATTCTTGAACCAGAATGTTC (JAK2 forward - SEQ ID NO: 1) and TTCTCACAAGCATTTGGTTTTGAAT (JAK2 reverse SEQ ID NO: 2) and as probes: VIC- CTCCACAGACACAGAC-MGB for JAK2WT (SEQ ID NO: 3) and 6-FAM- TCTCCACAGAAACAGAGA-MGB for Jak2 Vf ' m (SEQ ID NO: 4)
- Mass spectrometry analysis Size-exclusion chromatography of microvesicles was then performed in order to separate microvesicles from soluble proteins. Successive aliquot of 150 pL were collected and measurement of protein absorbance was performed. Fractions contained in tubes 6 to 11, containing microvesicles, were selected and then centrifuged at 20 500g for two hours. To finish, microvesicles were lysed using a 1% triton buffer.
- proteins were precipitated overnight at -20°C with 0.1 mol/L Ammonium Acetate glacial in 80% methanol (buffer 1). After centrifugation at 14000xg and 4°C for 15 min, the resulting pellets were washed twice with 100 pL of buffer 1 and further dried under vacuum (Speed-Vac concentrator). Proteins were then reduced by incubation with 10 pL of 5 mmol/L dithiotreitol (DTT) at 57°C for one hour and alkylated with 2 pL of 55 mmol/L iodoacetamide for 30 min at room temperature in the dark.
- DTT dithiotreitol
- Trypsin/LysC (Promega) was added twice at 1 : 100 (wt:wt) enzyme: substrate, at 37°C for 2 hrs first and then overnight. Samples were then loaded onto a homemade Cl 8 StageTips for desalting. Peptides were eluted using 40/60 MeCN/H20 + 0.1% formic acid and vacuum concentrated to dryness.
- the top 20 intense ions were subjected to Orbitrap for further fragmentation via high energy collision dissociation (HCD) activation and a resolution of 15 000 with the intensity threshold kept at 1.3 x 105.
- HCD high energy collision dissociation
- NCE Normalized collision energy
- the label free quantification was performed by peptide Extracted Ion Chromatograms (XICs) computed with MassChroQ version 2.2 (59).
- XICs peptide Extracted Ion Chromatograms
- MassChroQ version 2.2 59
- protein quantification XICs from proteotypic peptides shared between compared conditions (TopN matching) with two-missed cleavages were used. Median and scale normalization was applied on the total signal to correct the XICs for each biological replicate. To estimate the significance of the change in protein abundance, a linear model (adjusted on peptides and biological replicates) was performed and p-values were adjusted with a Benjamini-Hochberg FDR procedure with a control threshold set to 0.05.
- the mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE (60) partner repository with the dataset identifier PXD014451.
- Erythrocyte-derived microvesicles generated as mentioned above were centrifuged at 20 500g for 2 hrs, and then lysed in 100 DL RIPA buffer containing 150 mmol/L NaCl, 50 mmol/L TrisHCl, pH 7.4, 2 mmol/L EDTA, 0.5% sodium deoxycholate, 0.2% sodium dodecyl sulfate, 2 mmol/L activated orthovanadate, complete protease inhibitor cocktail tablet (Complet mini, Roche, France) and complete phosphatase inhibitor cocktail tablet (Roche, France). Protein content was quantified using the Micro BCA Protein Assay Kit (Thermo Scientific). Equal loading was checked using Ponceau red solution.
- Membranes were incubated with primary antibodies (1/1000) (Anti-GP91, BD611415; Anti-GSTTl, Abcaml99337; AntiMPO, Abcam45977). After secondary antibody incubation (anti-rat, Cell Signaling, 1/1000; anti rabbit or anti-mouse, Amersham, GE Healthcare, UK 1/3000), immunodetection was performed using an enhanced chemiluminescence kit (Immun-Star Western C kit, Bio-Rad). Bands were revealed using the LAS-4000 imaging system. Values reported from Western blots were obtained by band density analysis using with ImageJ software and expressed as the ratio protein of interest compared to Ponceau.
- Erythrocyte-derived microvesicles were stained with PKH26 dye (Sigma Aldrich) diluted in PBS, following manufacturer’s instructions, washed in PBS and then centrifuged at 20500g for 2 hrs. The 20500g supernatant served for control experiments.
- Murine endothelial cells (the cell line called SVEC4-10, CRL-2181, lot 70008729, ATCC) were then incubated with these stained microvesicles or an equal volume of the 20500g supernatant. After 2 hrs at 37°C, cells were washed 3 times with DMEM (Gibco).
- Cells were then fixed in PFA 4% for 5 min, and then costained with DAPI (0.1 Dg/mL, Sigma) in order to identify cell nuclei. Images were acquired using a Confocal microscope, Leica SP8 at 600 X magnification.
- Jak2 V617F is present in both hematopoietic and endothelial cells in patients with MPN (2, 3).
- VE-Cadherin being expressed during early embryonic life in a precursor of both endothelial and hematopoietic cells (13), Jak2 V6I7F Flex/WT; VE-Cadherin-cre, thereafter referred to as Jak2 V6I7F HC-EC, developed as expected a MPN, attested by higher spleen weight (2.3 to 5.7 % of body weight vs. 0.3 to 0.6 % for littermate controls, pO.OOOl), and higher haemoglobin level, platelet and white blood cell counts than littermate controls ( Figures 1A-D).
- mice To determine if this increased arterial contraction was due to Jak2 V617F in endothelial cells or in hematopoietic cells, we first generated mice expressing Jak2 V6I7F only in endothelial cells.
- Jak2 V6I7F Flex/WT mice We crossed Jak2 V6I7F Flex/WT mice with inducible VE-Cadherin-cre-ERT2 mice expressing the ere recombinase after tamoxifen injection only in endothelial cells.
- Jak2 V6I7F Flex/WT; VE-Cadherin-cre-ERT2 (thereafter referred to as ./ak2 i 6l / ' EC) mice did not develop MPN ( Figures 2A-D).
- mice expressing Jak2 V617F only in hematopoietic cells, by transplanting lethally irradiated C57BL/6 mice with Jak2 V617F bone marrow cells obtained from Jak2 V617F HC-EC mice. Irradiated C56BL/6 mice transplanted with Jak2WT BM were used as controls. Hematopoietic expression of Jak2 V617F induced a MPN ( Figures 2F-I) and an increased arterial response to phenylephrine (Figure 2J).
- microvesicles To investigate the interaction of erythrocyte-derived microvesicles with endothelial cells, we labeled microvesicles with the fluorescent dye PKH-26, incubated them with endothelial cells and then performed confocal microscopy on endothelial cells. Fluorescence was detected in endothelial cells, suggesting that erythrocyte-derived microvesicles were taken-up by endothelial cells (17). No difference in uptake was observed between microvesicles from Jak2 V617F HC-EC erythrocytes and their wild-type counterparts (Figure 3H).
- NOX2 cytochrome b-245 heavy and light chain
- Jak2 V6I7F erythrocyte-derived microvesicles carry MPO that confers a pro-oxidant phenotype to endothelial cells, leading to increased arterial contraction observed in Jak2 V6]7F ⁇ LC-EC mice.
- simvastatin also improves endothelial function through NO pathway and by preventing oxidative stress damage (23, 24).
- simvastatin also improves endothelial function through NO pathway and by preventing oxidative stress damage (23, 24).
- Fourteen days of treatment with simvastatin did not change spleen weight, haemoglobin level or platelet count (Figures 6 K-M). There was only a slight decrease in white blood cells count following simvastatin treatment (Figure 6N).
- simvastatin significantly improved aortic response to phenylephrine as compared to vehicle ( Figure 60).
- MPO is a polycationic heme-containing glycoprotein stored mainly in the azurophilic granules of neutrophils, but up to 30% of total cellular MPO can be released as active enzyme into the extracellular space.
- extra-cellular MPO can bind to red blood cells membrane and favours endothelial dysfunction in the context of ischemic heart disease (41-45).
- Our results demonstrate that MPO binds to erythrocyte-derived microvesicles, increases endothelial oxidative stress and vascular response to vasoconstrictors.
- Teofili L et al. Endothelial progenitor cells are clonal and exhibit the JAK2(V617F) mutation in a subset of thrombotic patients with Ph-negative myeloproliferative neoplasms. Blood 2011 ; 1 17(9):2700-2707.
- Musolino C et al Changes in advanced oxidation protein products, advanced glycation end products, and s-nitrosylated proteins, in patients affected by polycythemia vera and essential thrombocythemia. Clin. Biochem. 2012;45(16-17): 1439-1443.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Immunology (AREA)
- Molecular Biology (AREA)
- Analytical Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Medicinal Chemistry (AREA)
- Zoology (AREA)
- Organic Chemistry (AREA)
- Physics & Mathematics (AREA)
- Public Health (AREA)
- Biotechnology (AREA)
- Wood Science & Technology (AREA)
- Veterinary Medicine (AREA)
- Biochemistry (AREA)
- Pathology (AREA)
- Microbiology (AREA)
- Genetics & Genomics (AREA)
- Cell Biology (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Biomedical Technology (AREA)
- Hematology (AREA)
- Urology & Nephrology (AREA)
- Pharmacology & Pharmacy (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Physics & Mathematics (AREA)
- Food Science & Technology (AREA)
- Virology (AREA)
- Tropical Medicine & Parasitology (AREA)
- General Engineering & Computer Science (AREA)
- Biophysics (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Arterial cardiovascular events, i.e. the leading cause of death in patients with JAK2 V617F myeloproliferative neoplasms (MPN), are poorly understood. The inventors demonstrated that Jak2 V617F mice display a strong increase in arterial contraction with disturbed endothelial nitric oxide pathway and increased endothelial oxidative stress. This augmented arterial contraction was reproduced by circulating microvesicles isolated from patients carrying JAK2 V617F and by microvesicles derived from JAK2 V617F erythrocytes. Using proteomics, the inventors identified a high expression of myeloperoxidase in microvesicles derived from JAK2 V617F erythrocytes that could account for this effect. To assess the role of myeloperoxidase in this effect, the inventors then directly inhibited myeloperoxidase in microvesicles derived from Jak2 V617F erythrocytes and observed that it completely reversed the increase in endothelial oxidative stress induced by microvesicles derived from Jak2 V617F .The results prompt the inventors to conclude that JAK2 V617F MPN induce a potent increase in arterial contraction with increased endothelial oxidative stress, mediated by erythrocytes microvesicles and that myeloperoxidase (MPO) inhibitors would be suitable for preventing cardiovascular diseases in patient suffering from MPN.
Description
USE OF MYELOPEROXIDASE INHIBITORS FOR THE TREATMENT OF CARDIOVASCULAR DISEASES IN PATIENTS SUFFERING FROM MYELOPROLIFERATIVE NEOPLASMS
FIELD OF THE INVENTION:
The present invention is in the field of medicine.
BACKGROUND OF THE INVENTION:
Bcr/Abl-negative myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell disorders characterized by the proliferation of particular hematopoietic lineages without blockage in cell maturation. They include polycythemia vera, essential thrombocythemia, and primary myelofibrosis (1). JAK2 is the most common MPN driver gene. JAK2V617F is a gain of function mutation leading to growth factors hypersensitivity, detected in around 70% of MPNs (95% in polycythemia vera and 50% to 60% in essential thrombocythemia and pre-primary myelofibrosis / primary myelofibrosis) (1). JAK2V617F appears in pluripotent hematopoietic progenitor cells and is present in all erythroid and myeloid lineages (1). In addition, several groups described JAK2V617F in endothelial cells in the liver and the spleen of patients with splanchnic vein thrombosis (2, 3) and in circulating endothelial progenitor cells (4 6).
Cardiovascular diseases (CVD) reveal MPNs in about 30% of the patients and are the first cause of morbidity and mortality in these patients (7). Arterial events represent 60-70% of these cardio-vascular events (7). Interestingly, myocardial infarction without significant coronary stenosis by angiography was observed in 21% of patients with MNP (8) versus only 3% in a similar population without MPN (9). This observation prompted the European society of cardiology to recommend searching for MPNs in case of myocardial infarction without obstructive coronary artery disease (10). The mechanism underlying this link between myocardial infarction without obstructive coronary artery disease and MPNs is unknown, but vasoactive phenomenon (local intense vasoconstriction) can be suspected (11, 12).
SUMMARY OF THE INVENTION:
As defined by the claims, the present invention relates to the use of myeloperoxidase inhibitors for the treatment of cardiovascular diseases in patients suffering from myeloproliferative neoplasms.
DETAILED DESCRIPTION OF THE INVENTION:
Arterial cardiovascular events, i.e. the leading cause of death in patients with JAK2V617F myeloproliferative neoplasms (MPN), are poorly understood. The inventors demonstrated that Jak2V617F mice display a strong increase in arterial contraction with disturbed endothelial nitric oxide pathway and increased endothelial oxidative stress. This augmented arterial contraction was reproduced by circulating microvesicles isolated from patients carrying JAK2V617F zhά by microvesicles derived from JAK2V617F erythrocytes. Using proteomics, the inventors identified a high expression of myeloperoxidase in microvesicles derived from JAK2V617F erythrocytes that could account for this effect. To assess the role of myeloperoxidase in this effect, the inventors then directly inhibited myeloperoxidase in microvesicles derived from Jak2V6I7F erythrocytes and observed that it completely reversed the increase in endothelial oxidative stress induced by microvesicles derived from Jak2V6I7F. The results prompt the inventors to conclude that JAK2V6I7F MPN induce a potent increase in arterial contraction with increased endothelial oxidative stress, mediated by erythrocytes microvesicles and that myeloperoxidase (MPO) inhibitors would be suitable for preventing cardiovascular diseases in patient suffering from MPN.
Accordingly, the first object of the present invention relates to a method of preventing the occurrence of cardiovascular event in a patient suffering from a myeloproliferative neoplasm comprising administering to the patient a therapeutically effective amount of a myeloperoxidase inhibitor.
As used herein, the term“myeloproliferative neoplasm” or“MPN” has its general meaning in the art and refers to an acquired clonal hematopoietic stem cell disorder, characterized by an increase in one or more myeloid lineages. MPNs typically include polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF). They are a diverse but inter-related group of clonal disorders of pluripotent hematopoietic stem cells that share a range of biological, pathological and clinical features including the relative overproduction of one or more cell types from myeloid origin with growth factor independency/hypersensitivity, marrow hypercellularity, extramedullary hematopoiesis, spleno- and hepatomegaly, and thrombotic and/or hemorrhagic diathesis. An international working group for myeloproliferative neoplasms research and treatment (IWG-MRT) has been established to delineate and define these conditions (see for instance Vannucchi et al, CA Cancer J. Clin., 2009, 59: 171-191), and those disease definitions are to be applied for purposes of this specification.
In some embodiments, the patient harbours one mutation in JAK2. As used herein the term“JAK2” has its general meaning in the art and refers to the Janus Kinase 2 protein. The amino acid sequence of human JAK2 is well known in the art. Human JAK2 sequences are, for example, represented in the NCBI database (www.ncbi.orgwww.ncbi.nlm.nih.gov/), for example, under accession number NP 004963. Typical MPD associated mutation is the Jak2V6I7F mutation which refers to the point mutation (1849 G for T) in exon 14, which causes the substitution of phenylalanine for valine at codon 617 in the JAK homology JH2 domain. Other examples of JAK2 mutations include exon 12 mutations which can be substitutions, deletions, insertions and duplications, and all occur within a 44 nucleotides region in the JAK2 gene which encompasses amino acids 533-547 at the protein level. The most commonly reported mutations are small in-frame deletions of 3-12 nucleotides with a six nucleotides deletion being the most frequent. Complex mutations are present in one-third of cases with some mutations occurring outside this hotspot region. The N542-E543del is the most common mutation (23-30%), the K537L, E543-D544del and F537-K39delinsL represent 10-14%, and R541-E543delinsK comprise less than 10% of these mutations. JAK2 exon 12 mutations are located in a region close to the pseudo-kinase domain which acts as a linker between this domain and the Src homology 2 domain of JAK2.
As used herein, the term "cardiovascular event" as used herein refers to any disorder of the cardiovascular system including preferably any acute cardiovascular event. Acute cardiovascular events are, preferably, stable angina pectoris (SAP) or acute coronary syndrome (ACS). ACS patients can show unstable angina pectoris (UAP) or myocardial infarction (MI). MI can be an ST-elevation MI (STEMI) or a non-ST-elevation MI (NSTEMI). NSTE-ACS as used herein encompasses UAP and NSTEMI. The occurring of an MI can be followed by a left ventricular dysfunction (LVD), development of heart failure or even mortality. Further preferred cardiovascular events encompass cardiac brady- or tachyarrhythmias including sudden cardiac death and stroke (cerebrovascular events or accidents). Also, mortality can also refer to the death rate or the ratio of number of deaths to a given population of subjects.
As used herein the term“Myeloperoxidase” or MPO has its general meaning in the art and refers to a heme-containing enzyme. The enzyme uses hydrogen peroxide to oxidize chloride to hypochlorous acid. Other halides and pseudohalides (like thiocyanate) are also physiological substrates to MPO.
As used herein, a“MPO inhibitor” refers to any compound natural or not which is capable of inhibiting the activity of MPO, in particular MPO kinase activity. MPO inhibitors are well known in the art. The term encompasses any MPO inhibitor that is currently known in
the art or that will be identified in the future, and includes any chemical entity that, upon administration to a patient, results in inhibition or down-regulation of a biological activity associated with activation of the MPO. The term also encompasses inhibitor of expression. The MPO inhibition of the compounds may be determined using various methods well known in the art.
Examples of compounds that can be used as MPO-inhibitors are compounds described in WO 2006/062465, WO 2006/062465, WO 2003/089430, WO 2003/089430, or WO 2003/089430.
In some embodiments, the MPO inhibitor of the present invention is selected from the group consisting of:
l-butyl-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
l-isobutyl-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4-one;
l-(pyridin-2-ylmethyl)-2-thioxo- 1,2,3, 5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4- one;
l-(2-fluoro-benzyl)-2-thioxo- 1,2,3, 5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4- one;
l-[2-(2-methoxyethoxy)-3-propoxybenzyl]-2-thioxo-l,2,3,5-tetrahydro- pyrrolo[3,2-d]pyrimidin-4-one;
l-(6-ethoxy-pyridin-2-ylmethyl)-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2- d]pyrimidin-4-one;
1 -piperidin-3 -ylmethyl-2-thioxo- 1 ,2,3 , 5 -tetrahydro-pyrrolo[3 ,2-d]pyrimidin-4- one;
l-butyl-4-thioxo-l,3,4,5-tetrahydro-2H-pyrrolo[3,2-d]pyrimidin-2-one;
l-(2-isopropoxyethyl)-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-4- one;
1 -(2-methoxy-2-methylpropyl)-2-thioxo- 1 ,2,3 , 5 -tetrahydro-pyrrolo[3 ,2- d]pyrimidin-4-one;
l-(2-ethoxy-2-methylpropyl)-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2- d]pyrimidin-4-one;
l-(piperi din-4-ylmethyl)-2-thioxo- 1,2,3, 5-tetrahydro-pyrrolo[3,2-d]pyrimidin- 4-one;
l-[(l-methylpiperidin-3-yl)methyl]-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2- d]pyrimidin-4-one;
l-[2-hydroxy-2-(4-methoxyphenyl)ethyl]-2-thioxo- 1,2,3, 5-tetrahydro- pyrrolo[3,2-d]pyrimidin-4-one;
1 -(2-methoxybenzyl)-2-thioxo- 1 ,2,3 , 5-tetrahydro-pyrrolo [3 ,2-d]pyrimidin-4- one;
1 -(3 -methoxybenzyl)-2-thioxo- 1 ,2,3 , 5-tetrahydro-pyrrolo[3 ,2-d]pyrimidin-4- one;
l-(2,4-dimethoxybenzyl)-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2-d]pyrimidin-
4-one;
l-[(3-chloropyridin-2-yl)methyl]-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2- d]pyrimidin-4-one;
l-{ [3-(2-ethoxyethoxy)pyridin-2-yl]methyl}-2 -thioxo- 1,2, 3, 5-tetrahydro- pyrrolo[3,2-d]pyrimidin-4-one;
l-[(6-oxo-l,6-dihydropyridin-2-yl)methyl]-2-thioxo-l,2,3,5-tetrahydro- pyrrolo[3,2-d]pyrimidin-4-one;
l-(lH-indol-3-ylmethyl)-2 -thioxo- 1,2,3, 5-tetrahydro-pyrrolo[3,2-d]pyrimidin- 4-one;
1 -( 1 H-benzimidazol-2-ylmethyl)-2-thioxo- 1 ,2,3 , 5 -tetrahydro-pyrrolo[3 ,2- d]pyrimidin-4-one;
l-[(5-chloro-lH-indol-2-yl)methyl]-2 -thioxo- 1,2,3, 5-tetrahydro-pyrrolo[3, 2- d]pyrimidin-4-one;
1 - [(5 -fluoro- 1 H-indol-2-yl)methyl] -2-thioxo- 1,2,3 , 5-tetrahydro-pyrrolo[3 ,2- d]pyrimidin-4-one;
l-(lH-indol-6-ylmethyl)-2 -thioxo- 1,2,3, 5-tetrahydro-pyrrolo[3,2-d]pyrimidin- 4-one;
l-(lH-indol-5-ylmethyl)-2 -thioxo- 1,2,3, 5-tetrahydro-pyrrolo[3,2-d]pyrimidin- 4-one;
1 - [(5 -fluoro- 1 H-indol-3 -yl)methyl] -2-thioxo- 1,2,3 , 5-tetrahydro-pyrrolo[3 ,2- d]pyrimidin-4-one;
l-(lH-imidazol-5-ylmethyl)-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2- d]pyrimidin-4-one;
l-(lH-imidazol-2-ylmethyl)-2-thioxo-l,2,3,5-tetrahydro-pyrrolo[3,2- d]pyrimidin-4-one;
l-[(5-chloro-lH-benzimidazol-2-yl)methyl]-2-thioxo- 1,2,3, 5-tetrahydro- pyrrolo[3,2-d]pyrimidin-4-one;
1 - [(4, 5 -dimethyl- 1 H-benzimidazol-2-yl)methyl] -2-thioxo- 1 ,2,3 , 5-tetrahydro- pyrrolo[3,2-d]pyrimidin-4-one;
7-bromo- 1 -isobutyl-2 -thioxo- 1 ,2,3 ,5-tetrahydro-pyrrolo[3 ,2-d]pyrimidin-4- one; and
1 -(3 -chlorophenyl)-2-thioxo- 1,2,3 , 5-tetrahydro-pyrrolo[3 ,2-c]pyrimidin-4-one.
1.3-diisobutyl-8-methyl-6-thioxanthine;
1.3-dibutyl-8-methyl-6-thioxanthine;
3-isobutyl- 1 , 8-dimethyl-6-thioxanthine;
3-(2-methylbutyl)-6-thioxanthine;
3-isobutyl-8-methyl-6-thioxanthine;
3-isobutyl-2-thioxanthine;
3-isobutyl-2,6-dithioxanthine;
3-isobutyl-8-methyl-2-thioxanthine;
3-isobutyl-7-methyl-2-thioxanthine;
3-cyclohexylmethyl-2-thioxanthine;
3-(3-methoxypropyl)-2-thioxanthine;
3-cyclopropylmethyl-2-thioxanthine;
3-isobutyl- 1 -methyl-2-thioxanthine;
3-(2-tetrahydrofuryl-methyl)-2-thioxanthine;
3-(2-methoxy-ethyl)-2-thioxanthine;
3 -(3 -( 1 -morpholinyl)-propyl)-2-thioxanthine;
3-(2-furyl-methyl)-2-thioxanthine;
3-(4-methoxybenzyl)-2-thioxanthine;
3-(4-fluorobenzyl)-2-thioxanthine;
3 -phenethyl-2-thioxanthine;
(+)-3 -(2-tetrahydrofuryl-methyl)-2-thioxanthine;
(-)-3-(2-tetrahydrofuryl-methyl)-2-thioxanthine; and
3 -n-butyl-2-thioxanthine .
3-(pyridin-2-ylmethyl)-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one;
3-(pyridin-3-ylmethyl)-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one;
3-(pyridin-4-ylmethyl)-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one;
3-{[3-ethoxy-4-(2-ethoxyethoxy)pyridin-2-yl]methyl}-2-thioxo-l,2,3,7- tetrahydro-6H-purin-6-one;
3-[(5-fhioro-lH-indol-2-yl)methyl]-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6- one;
3-[(5-fluoro-lH-indol-2-yl)methyl]-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6- one;
3-[(2-butyl-4-chloro-lH-imidazol-5-yl)methyl]-2-thioxo-l,2,3,7-tetrahydro-
6H-purin-6-one;
3-(lH-benzimidazol-2-ylmethyl)-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one; 3-[l-(lH-benzimidazol-2-yl)ethyl]-2-thioxo- 1,2,3, 7-tetrahydro-6H-purin-6- one;
3-[(5-chloro-lH-indol-3-yl)methyl]-2 -thioxo- 1,2,3, 7-tetrahydro-6H-purin-6- one
3-[(4-fluoro-lH-indol-3-yl)methyl]-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6- one;
3 - [2-( 1 H-Benzimidazol-2-yl)ethyl] -2-thioxo- 1 ,2,3 , 7 -tetrahydro-6H-purin-6- one;
3 -( 1 H-Pyrazol-3 -ylmethyl)-2-thioxo- 1,2,3 , 7-tetrahydro-6H-purin-6-one;
3-[(5-Methylpyrazin-2-yl)methyl)-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one; 3 - [(3 -Isopropyli soxazol-5 -yl)methyl] -2-thioxo- 1 ,2,3 , 7-tetrahydro-6H-purin-6- one;
3-[(4-Methyl-l,2,5-oxadiazol-3-yl)methyl]-2-thioxo-l,2,3,7-tetrahydro-6H- purin-6-one;
3-[(6-Butoxypyridin-2-yl)methyl]-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one;
3-[(4-Butoxypyridin-2-yl)methyl]-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one;
3-[(3-Butoxypyridin-2-yl)methyl]-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-(Pyridin-2-ylmethoxy)propyl]-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6- one;
3-[(3,5-Dimethylisoxazol-4-yl)methyl]-2-thioxo-l,2,3,7-tetrahydro-6H-purin-
6-one;
3-[(l -Methyl- lH-indol-2-yl)methyl]-2-thioxo- 1,2,3, 7-tetrahydro-6H-purin-6- one;
3-(2-Phenyl-2-pyridin-2-ylethyl)-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one;
3-(Quinolin-4-ylmethyl)-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one;
3-[(6-Phenoxypyridin-3-yl)methyl]-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6- one;
3-{2-[(Quinolin-4-ylmethyl)amino]ethyl}-2-thioxo-l,2,3,7-tetrahydro-6H- purin-6-one;
3 -(2- { [( 1 -Methyl- 1 H-indol-3 -yl)methyl]amino } ethyl)-2-thioxo- 1 ,2,3 , 7- tetrahydro-6H-purin-6-one;
3-{2-[Methyl(quinolin-4-ylmethyl)amino]ethyl}-2 -thioxo- 1,2, 3, 7-tetrahydro- 6H-purin-6-one;
3-(2-Aminopropyl)-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one
trifluoroacetate;
3-{2-[(Pyridin-2-ylmethyl)amino]propyl}-2-thioxo- 1,2,3, 7-tetrahydro-6H- purin-6-one trifluoroacetate;
3-{2-[(Pyridin-3-ylmethyl)amino]propyl}-2-thioxo- 1,2,3, 7-tetrahydro-6H- purin-6-one;
3-{2-[(Pyridin-4-ylmethyl)amino]propyl}-2-thioxo- 1,2,3, 7-tetrahydro-6H- purin-6-one;
3-(2-{[(6-Chloropyridin-3-yl)methyl]amino}propyl)-2-thioxo-l,2,3,7- tetrahydro-6H-purin-6-one trifluoroacetate;
3 -[2-({ [6-(Trifluoromethyl)pyri din-3 -yl] methyl }amino)propyl] -2-thioxo-
1.2.3.7-tetrahydro-6H-purin-6-one trifluoroacetate;
3-(2-{[(4,6-Dichloropyrimidin-5-yl)methyl]amino}propyl)-2-thioxo-l,2,3,7- tetrahydro-6H-purin-6-one;
3-[2-({[2-(Dimethylamino)pyrimidin-5-yl]methyl}amino)propyl]-2-thioxo-
1.2.3.7-tetrahydro-6H-purin-6-one;
3-{2-[(Quinolin-2-ylmethyl)amino]propyl}-2-thioxo- 1,2,3, 7-tetrahydro-6H- purin-6-one trifluoroacetate;
3-{2-[(Quinolin-3-ylmethyl)amino]propyl}-2-thioxo- 1,2,3, 7-tetrahydro-6H- purin-6-one;
3-(2-{[(l-tert-Butyl-3,5-dimethyl-lH-pyrazol-4-yl)methyl]amino}propyl)-2- thioxo-l,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-({[l-(l,l-Dioxidotetrahydro-3-thienyl)-3,5-dimethyl-lH-pyrazol-4- yl]methyl }amino)propyl]-2-thioxo- 1 ,2,3,7-tetrahydro-6H-purin-6-one;
3-{2-[(lH-Benzoimidazol-2-ylmethyl)amino]propyl}-2 -thioxo- 1,2, 3,7- tetrahydro-6H-purin-6-one;
3-[2-({[l-(Phenylsulfonyl)-lH-pyrrol-2-yl]methyl}amino]propyl]-2-thioxo-
1.2.3.7-tetrahydro-6H-purin-6-one trifluoroacetate;
3-{2-[({ l-[(4-methylphenyl)sulfonyl]-lH-pyrrol-2-yl}methyl)amino]propyl}-
2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one trifluoroacetate;
3 -(2- { [( 1 -methyl- 1 H-py rrol-2-yl)methyl]amino } propyl)-2-thioxo- 1 ,2,3 , 7- tetrahydro-6H-purin-6-one;
3 - [2-( { [ 1 -(4-sec-Butylphenyl)- 1 H-pyrrol-2-yl] methyl } amino)propyl)-2-thioxo- l,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-({[l-(3 -Methoxyphenyl)- lH-pyrrol-2-yl] methyl } amino)propyl] -2-thioxo- l,2,3,7-tetrahydro-6H-purin-6-one;
3 - [2-( { [2, 5-Dimethyl- 1 -( 1 , 3 -thiazol-2-yl)- lH-pyrrol-3 - yl]methyl}amino)propyl]-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one;
3-[2-({[4-(3-Chlorobenzoyl)-l-methyl-lH-pyrrol-2-yl]methyl}amino)propyl]-
2-thioxo- 1,2,3 , 7-tetrahydro-6H-purin-6-one;
3-{2-[(lH-Imidazol-2-ylmethyl)amino]propyl}-2-thioxo-l,2,3,7-tetrahydro- 6H-purin-6-one;
3-(2-{ [(1 -Methyl- lH-imidazol-2-yl)methyl]amino}propyl)-2-thioxo-l, 2,3,7- tetrahydro-6H-purin-6-one;
3-(2-{ [(4-Bromo-l -methyl- lH-imi dazol-5-yl)methyl]amino}propyl)-2 -thioxo- l,2,3,7-tetrahydro-6H-purin-6-one;
3-(2-{ [(1 -Methyl- lH-indol-3-yl)methyl]amino}propyl)-2-thioxo- 1,2, 3,7- tetrahydro-6H-purin-6-one;
2-Thioxo-3-{2-[(lH-l,2,3-triazol-5-ylmethyl)amino]propyl}-l,2,3,7- tetrahydro-6H-purin-6-one;
3-[2-({[l-(Benzyloxy)-lH-imidazol-2-yl]methyl}amino)propyl]-2-thioxo- l,2,3,7-tetrahydro-6H-purin-6-one;
3-(2-{[(6-Bromo-2-methylimidazo[l,2-a]pyridin-3-yl)methyl]amino}propyl}-
2-thioxo- 1,2,3 , 7-tetrahydro-6H-purin-6-one;
3-{2-[({ l-[2-(2-Methoxyphenoxy)ethyl]-lH-pyrrol-2- yl}methyl)amino]propyl]-2-thioxo-l,2,3,7-tetrahydro-6H-purin-6-one;
N-[ 1 -Methyl-2-(6-oxo-2-thioxo- 1 ,2,6,7-tetrahydro-3H-purin-3- yl)ethyl]pyridine-2-carboxamide;
N-[ 1 -Methyl-2-(6-oxo-2-thioxo- 1 ,2,6,7-tetrahydro-3H-purin-3- yl)ethyl]nicotinamide;
N-[ 1 -Methyl-2-(6-oxo-2-thioxo- 1 ,2,6,7-tetrahydro-3H-purin-3-yl)- ethyl ] i soni cotinamide;
N-[l-methyl-2-(6-oxo-2-thioxo-l,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]-l,8- naphthyridine-2-carboxamide;
N-[ 1 -Methyl-2-(6-oxo-2-thioxo- 1 ,2,6,7-tetrahydro-3H-purin-3- yl)ethyl]quinoline-2-carboxamide;
N-[ 1 -Methyl-2-(6-oxo-2-thioxo- 1 ,2,6,7-tetrahydro-3H-purin-3- yl)ethyl]pyrimidine-2-carboxamide; and
N-[ 1 -Methyl-2-(6-oxo-2-thioxo- 1 ,2,6,7-tetrahydro-3H-purin-3-yl)ethyl]- 1H- imidazole-2-carboxamide trifluroaceate.
In some embodiment, the MPO inhibitor is AZD5904 which has the formula of:
In some embodiments, the MPO inhibitor is an inhibitor of MPO expression. An “inhibitor of expression” refers to a natural or synthetic compound that has a biological effect to inhibit the expression of a gene. In a preferred embodiment of the invention, said inhibitor of gene expression is a siRNA, an antisense oligonucleotide or a ribozyme. For example, anti- sense oligonucleotides, including anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of MPO mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of MPO, 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 encoding MPO 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). Small inhibitory RNAs (siRNAs) can also function as inhibitors of expression for use in the present invention. MPO gene expression can be reduced by contacting a subject or cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that MPO gene expression is specifically inhibited (i.e. RNA interference or RNAi). Antisense
oligonucleotides, siRNAs, shRNAs and ribozymes 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 cells expressing MPO. 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; SV40-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.
By a "therapeutically effective amount" of the inhibitor as above described is meant a sufficient amount to provide a therapeutic effect. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed, the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide employed; and like factors well known in the medical arts. For example, it is well within the skill of the art to start doses of the compound at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. However, the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day. Typically, the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject to be treated. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from 1 mg to about 100 mg of the active ingredient. An effective amount of the drug
is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
Typically, the inhibitor of the present invention is combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form pharmaceutical compositions. "Pharmaceutically" or "pharmaceutically acceptable" refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate. 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. 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 inhibitor at 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.
A further object of the present invention relates to a method of determining whether a patient suffering from a myeloproliferative neoplasm is at risk of having a cardiovascular event comprising the steps of determining the level of microvesicles derived from Jak2V6!7F erythrocytes in a blood sample obtained from the patient wherein the level correlates with the risk of having a cardiovascular disease.
As used herein, the term“risk" relates to the probability that an event will occur over a specific time period, as in the conversion to a cardiovascular event, and can mean a subject's "absolute" risk or "relative" risk. Absolute risk can be measured with reference to either actual observation post-measurement for the relevant time cohort, or with reference to index values developed from statistically valid historical cohorts that have been followed for the relevant time period. Relative risk refers to the ratio of absolute risks of a subject compared either to the absolute risks of low risk cohorts or an average population risk, which can vary by how clinical risk factors are assessed. Odds ratios, the proportion of positive events to negative events for a given test result, are also commonly used (odds are according to the formula p/(l-p) where p is the probability of event and (1- p) is the probability of no event) to no- conversion. Alternative continuous measures which may be assessed in the context of the present invention include time to a cardiovascular event conversion and therapeutic a cardiovascular event conversion risk reduction ratios.
Thus the expression "determining whether a patient is at risk of having a cardiovascular event" as used herein means that the patient to be analyzed by the method of the present invention is allocated either into the group of patients of a population having an elevated risk, or into a group having a reduced risk of having a cardiovascular event. An elevated risk as referred to in accordance with the present invention, preferably, means that the risk of developing a cardiovascular event within a predetermined predictive window is elevated significantly (i.e. increased significantly) for a patient with respect to the average risk for a cardiovascular event or cardiac mortality in a population of patients. A reduced risk as referred to in accordance with the present invention, preferably, means that the risk of developing a cardiovascular event within a predetermined predictive window is reduced significantly for a patient with respect to the average risk for a cardiovascular event or cardiac mortality in a population of patients. Particularly, a significant increase or reduction of a risk is an increase or reduction or a risk of a size which is considered to be significant for prognosis, particularly said increase or reduction is considered statistically significant. The terms "significant" and "statistically significant" are known by the person skilled in the art. Thus, whether an increase or reduction of a risk is significant or statistically significant can be determined without further ado by the person skilled in the art using various well known statistic evaluation tools.
As used herein the term“blood sample” means a whole blood, serum, or plasma sample obtained from the patient. Preferably the blood sample, according to the invention, is a plasma sample. A plasma sample may be obtained using methods well known in the art. For example, blood may be drawn from the patient following standard venipuncture procedure on tri-sodium
citrate buffer. Plasma may then be obtained from the blood sample following standard procedures including but not limited to, centrifuging the blood sample at about 2500*g for about 15 minutes (room temperature), followed by pipetting of the plasma layer. Platelet-free plasma (PFP) will be obtained following a second centrifugation at about 2500*g for 15 min. Analyses can be performed directly on this PFP. Alternatively, microvesicles may be more specifically isolated by further centrifuging the PFP at about 15,000 to about 25,000*g at 4°C. Different buffers may be considered appropriate for resuspending the pelleted cellular debris which contains the MPs. Such buffers include reagent grade (distilled or deionized) water and phosphate buffered saline (PBS) pH 7.4 or NaCl 0.9%. Preferably, PBS buffer (Sheath fluid) is used.
As used herein the term“microvesicle” or“MV” or“extracellular vesicles” has its general meaning in the art and denotes a plasma membrane vesicle shed from an apoptotic or activated cell. The size of microvesicles / extracellular vesicles ranges from 0.1 pm to 1 pm in diameter. The surface markers of microvesicles are the same as the cells from they originated.
Standard methods for determining the level of microvesicles in a blood sample are well known in the art and typically involve the methods described in the EXAMPLE.
In some embodiments, the level of the microvesicles derived from Jak2V617F erythrocytes is compared to a predetermined reference value. Typically, the predetermined reference value is a threshold value or a cut-off value. Typically, a "threshold value" or "cut off value" can be determined experimentally, empirically, or theoretically. A threshold value can also be arbitrarily selected based upon the existing experimental and/or clinical conditions, as would be recognized by a person of ordinary skilled in the art. For example, retrospective measurement of expression levels in properly banked historical patient samples may be used in establishing the predetermined reference value. The threshold value has to be determined in order to obtain the optimal sensitivity and specificity according to the function of the test and the benefit/risk balance (clinical consequences of false positive and false negative). Typically, the optimal sensitivity and specificity (and so the threshold value) can be determined using a Receiver Operating Characteristic (ROC) curve based on experimental data. For example, after quantifying the expression level in a group of reference, one can use algorithmic analysis for the statistic treatment of the determined levels in samples to be tested, and thus obtain a classification standard having significance for sample classification. The full name of ROC curve is Receiver Operator Characteristic Curve, which is also known as receiver operation characteristic curve. It is mainly used for clinical biochemical diagnostic tests. ROC curve is a comprehensive indicator that reflects the continuous variables of true positive rate (sensitivity)
and false positive rate (1-specificity). It reveals the relationship between sensitivity and specificity with the image composition method. A series of different cut-off values (thresholds or critical values, boundary values between normal and abnormal results of diagnostic test) are set as continuous variables to calculate a series of sensitivity and specificity values. Then sensitivity is used as the vertical coordinate and specificity is used as the horizontal coordinate to draw a curve. The higher the area under the curve (AUC), the higher the accuracy of diagnosis. On the ROC curve, the point closest to the far upper left of the coordinate diagram is a critical point having both high sensitivity and high specificity values. The AUC value of the ROC curve is between 1.0 and 0.5. When AUC>0.5, the diagnostic result gets better and better as AUC approaches 1. When AUC is between 0.5 and 0.7, the accuracy is low. When AUC is between 0.7 and 0.9, the accuracy is moderate. When AUC is higher than 0.9, the accuracy is quite high. This algorithmic method is preferably done with a computer. Existing software or systems in the art may be used for the drawing of the ROC curve, such as: MedCalc 9.2.0.1 medical statistical software, SPSS 9.0, ROCPOWER.SAS, DESIGNROC.FOR, MULTIREADER POWERSAS, CREATE-ROC.SAS, GB STAT VIO.O (Dynamic Microsystems, Inc. Silver Spring, Md., USA), etc.
In some embodiments, the predetermined reference value was established in a population of patients who did not have a cardiovascular event when blood was drawn. Accordingly when the level of microvesicles is higher than the predetermined reference value, it is concluded that the patient is at risk of having a cardiovascular event. On contrary, when the level of microvesicles is lower than the predetermined reference value, then is it concluded that the patient is not at risk of having a cardiovascular event.
In some embodiments, high statistical significance values (e.g. low P values) are obtained for a range of successive arbitrary quantification values, and not only for a single arbitrary quantification value. Thus, in some embodiments, instead of using a definite predetermined reference value, a range of values is provided. Therefore, a minimal statistical significance value (minimal threshold of significance, e.g. maximal threshold P value) is arbitrarily set and a range of a plurality of arbitrary quantification values for which the statistical significance value calculated at step g) is higher (more significant, e.g. lower P value) are retained, so that a range of quantification values is provided. This range of quantification values includes a "cut-off1 value as described above. For example, according to this specific embodiment of a "cut-off1 value, the outcome can be determined by comparing the expression level with the range of values which are identified. In some embodiments, a cut-off value thus consists of a range of quantification values, e.g. centered on the quantification value for which
the highest statistical significance value is found (e.g. generally the minimum p value which is found). For example, on a hypothetical scale of 1 to 10, if the ideal cut-off value (the value with the highest statistical significance) is 5, a suitable (exemplary) range may be from 4-6. For example, a patient may be assessed by comparing values obtained by determining the level of microvesicles, where values greater than 5 reveal that the patient is at risk of having a cardiovascular event and values less than 5 reveal that the patient is not at risk of having a cardiovascular event. In some embodiments, a patient may be assessed by comparing values obtained by measuring the level of microvesicles and comparing the values on a scale, where values above the range of 4-6 indicate that the patient is at risk of having a cardiovascular event and values below the range of 4-6 indicate that the patient is not at risk of having a cardiovascular event, with values falling within the range of 4-6 indicating an intermediate risk.
The result given by the method of the invention may be used as a guide in determining how frequently a cardiovascular event should be screened, in selecting a therapy or treatment regimen for the patient. For example, when the patient has been determined as having a high risk of a cardiovascular event, he can be eligible for a therapy with a MPO inhibitor as described herein, and/or with a statin. As used herein the term“Statin” has its general meaning in the art and refers to a class of drugs that are inhibitors of HMG-CoA reductase. Examples of statin include but are not limited to pravastatin, fluvastatin, atorvastatin, lovastatin, simvastatin, rosuvastatin, and cerivastatin. Statins may be in the form of a salt, hydrate, solvate, polymorph, or a co-crystal. Statins may also be in the form of a hydrate, solvate, polymorph, or a co-crystal of a salt. Statins may also be present in the free acid or acetone form.
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. Jak2V617F in hematopoietic and endothelial cells increases arterial contraction in an endothelial-dependent manner.
Representative picture of the spleen (A). Haemoglobin (B), platelet (C) and white blood cell count (D) of 8 to 12 weeks old control mice (Jak2WT, n=13) and Jak2V617F Flex/WT ;VE- Cadherin-cre mice ( Jak2V6I7F HC-EC, n=13). Cumulative dose-response curves to phenylephrine (E) (Jak2WT, n=13; Jak2V6I7F HC-EC, n=13), to angiotensin II (G) (Jak2WT, n=3; Jak2l '>!71 HC-EC, n=4) and contraction response to potassium chloride (80 mmol/L) (F) (Jak2WT, n=13; Jak2V6I7F HC-EC, n=13) of aortas with endothelium. Cumulative dose-
response curves to phenylephrine of aortas without endothelium (H) (Jak2WT, n=6; Jak2V617F HC-EC, n=6). Diameter change of femoral artery after phenylephrine injection (10-3 mol/L) (I) (Jak2WT, n=8; Jak2V6I7F HC-EC, n=8). Electrocardiogram recording before and after intravenous phenylephrine injection (3 mg/kg; Jak2WT, n=13; Jak2V6I7F HC-EC, n=6) (J), with representative images of the changes observed in 5/6 Jak2V617F HC-EC vs. only 4/13 Jak2WT (p=0.057) (K). Quantitative data are expressed as median with interquartile range and cumulative dose-response curves are expressed as mean with standard error of the mean. Abbreviations: * p<0.05, *** p< 0.001; ns, not significant. Cumulative dose response curves and electrocardiogram recording were compared using an analysis of variance for repeated measures and other data were compared using the Mann- Whitney U-test. All tests were 2 sided.
Figure 2. Jak2V617F specifically expressed in hematopoietic cells, but not in endothelial cells, increases arterial contraction
Representative picture of the spleen (A and F). Blood cell count of 10 to 13 weeks old control mice (Jak2WT, n=7) and .Iak21 6171 Flex/WT; VE-Cadherin-cre-ERT2 mice (, Jak2V617F EC, n=7) (B, C, D) and of 13 to 15 weeks old chimeric C57BL/6 mice transplanted with a bone marrow of wild-type mice (Jak2WT, n=5) or of.lak2l 7 /7/ HC-F.C mice (Jak2V617F HC, n=5) (G, H, I). Data are expressed as median with interquartile range. Cumulative dose-response curve to phenylephrine of aortas from Jak2WT (n=7) and Jak2V6!7F EC (n=7) (E) and from Jak2WT (n=5) and Jak2V6I7F HC (n=5) (J). Quantitative data are expressed as median with interquartile range and cumulative dose-response curves are expressed as mean with standard error of the mean. Abbreviations: * p<0.05, ** p< 0.01; ns, not significant. Cumulative dose response curves were compared using an analysis of variance for repeated measures and other data were compared using the Mann- Whitney U-test. All tests were 2 sided.
Figure 3. Microvesicles derived from Jak2V617F red blood cells are responsible for an increased arterial contraction
Cumulative dose-response curves to phenylephrine of aortas from WT mice incubated with microvesicles isolated from Jak2V617F patients (n=7) and controls (n=5) at their circulating concentration (A). Cumulative dose-response curves to phenylephrine of aortas from WT mice incubated with microvesicles generated from platelets (n=5 and n=5, respectively) (B), PBMC (n=5 and n=6, respectively) (C), PMNC (n=5 and n=5) (D) and red blood cells (n=9 and n=4, respectively) (E) from Jak2V617F HC-EC mice (Jak2V617F) or littermate control mice (WT). Change in the diameter of femoral artery induced by phenylephrine injection (10-3 mol/L) in control mice previously injected with control erythrocyte-derived microvesicles (JAK2WT MV GR WT, n=10) or with Jak2V617F erythrocyte-derived microvesicles (JAK2WT MV GR
Jak2V617F n=10) (F). Allelic discrimination plot of ARN isolated from microvesicles derived from Jak2\VT and Jak2V6!7F erythrocytes (n=3 per group) (no template control, black) (G). Quantification (H) of the uptake by endothelial cells (HUVEC) of erythrocyte-derived microvesicles from JAK2V617F mice (n=5) or JAK2m mice (n=5) or respective 20500 g supernatant (n=3 for each group). Cumulative dose-response curve to phenylephrine of aortas from WT mice injected with vehicle (n 5) or with epoietin (n=8) (I). Quantitative data are expressed as median with interquartile range and cumulative dose-response curves are expressed as mean with standard error of the mean. Abbreviations: *p<0.05; ** p< 0.01; EPO, epoietin, HUVEC, Human umbilical vein endothelial cells, MVs, microvesicles; ns, not significant; NTC, no template control; PBMC, peripheral blood mononuclear cells; PMNC, polymorphonuclear cells; RBC, Red blood cells; SNT, Supernatant; WT, wild type. Cumulative dose response curves were compared using an analysis of variance for repeated measures and other data were compared using the Mann- Whitney U-test. All tests were 2 sided
Figure 4. Disturbed endothelial NO pathway and increased oxidative stress status.
Cumulative dose-response curve of aortas from Jak2V617F Flex/WT ;VE-Cadherin-cre mice (Jak2V6I7F HC-EC mice) and littermate controls (Jak2WT) to acetylcholine (n=l l and n=l l, respectively) (A), to S-Mtroso-N-Acetylpenicillamine (SNAP) (n=5 and n=6, respectively) (C) and to phenylephrine after L-NAME incubation (n=l 1 and n=7, respectively) (E). Diameter change of femoral arteries after injection of acetylcholine (10-2 mol/L) (Jak2WT, n=8; Jak2V617F HC-EC, n=8 (B) and SNAP (10-3 mol/L) (Jak2WT, n=4, .VA2, 7,/7/ HC-EC, n=4) (D). Quantification of reactive oxygen species (ROS) generation per endothelial cell in: control mice (Jak2WT) vs. Jak2V617F HC-EC mice (F); control mice (Jak2WT) and Jak2i r'r/I Flex/WT : VE-Cadherin-cre-ERT2 mice (Jak2V6I7F EC) (G); control mice injected with microvesicles derived from control (JAK2WT MV GR WT, n=6) or Jak2V617F erythrocytes (JAK2WT MV GR Jak2V617F n=6) (H). Bar scale 10 pm. Cumulative dose-response curve to phenylephrine of aortas from Jak2V617F HC-EC mice treated with vehicle (n=5) and with NAC (n=7) (I). Quantitative data are expressed as median with interquartile range and compared using the Mann-Whitney U-test and cumulative dose-response curves are expressed as mean with standard error of the mean and compared using an analysis of variance for repeated measures. Abbreviations: * p<0.05, *** p< 0.001; CY24A, NAC, N-Acetyl-cysteine; ns, not significant.
Figure 5. Myeloperoxidase carried by erythrocyte-derived microvesicles from Jak2V617F mice is responsible for increased endothelial oxidative stress.
Volcano plot obtained by using quantitative label-free mass spectrometry analysis of proteins isolated from microvesicles derived from Jak2V6i7F (n=6) and JAK2WT (n=4)
erythrocytes (ratio JAK2V617FMAK2WT) (A), only proteins involved in cellular oxidant detoxification (GO 0098869) and ROS metabolic process (GO 0072593) are presented (line corresponds to p-value< 0.05). Respective quantification of GSTT1 (B) and MPO (C) western blots performed on erythrocyte-derived microvesicles (JAK2WT erythrocyte microvesicles, n=l l; Jak2V617F erythrocyte microvesicles, n=l l). Quantification of reactive oxygen species (ROS) generation per endothelial cell (HUVEC) after exposition of erythrocyte-derived microvesicles from control mice (Jak2WT) (n=4) and Jak2i >171 HC-EC mice and without (n=4) and with (n=4) preincubation with a myeloperoxidase inhibitor (MPOi, PF06281355, Sigma, 5 pmol/L) (D). Bar scale 10 pm. Quantitative data are expressed as median with interquartile range and compared using the Mann- Whitney U-test and Kruskal- Wallis test for multiple comparison. Abbreviations: * p<0.05, *** p< 0.001; CY24A, Cytochrome b-245 light chain; CY24B, Cytochrome-b245 Heavy chain; GSTT1, glutathione s transferase theta 1; MPO, myeloperoxidase; ns, not significant.
Figure 6. Simvastatin improves the increased arterial contraction induced by
Jak2V617F
Spleen to body weight ratio (A), haemoglobin level (B), platelet (C) and white blood cell count (D) in Jak2V6I7F Flex/WT ;VE-Cadherin-cre mice (Jak2V617F HC-EC) treated with vehicle {Jak2V617F HC-EC vehicle, n=4) or with hydroxyurea ( Jak2V6I7F HC-EC HU, n=7). Spleen to body weight ratio (F), haemoglobin level (G), platelet (H) and white blood cell count (I) in Jak2V617F HC-EC mice treated with vehicle (Jak2V6I7F HC-EC vehicle, n=5) or with ruxolitinib {Jak2V6I7F HC-EC ruxolitinib, n=4). Spleen to body weight ratio (K), haemoglobin level (L), platelet (M) and white blood cell count (N) in Jak2V6I7F HC-EC mice treated with vehicle (n=10) or with simvastatin ( Jak2V617F HC-EC simvastatin, n=7). Cumulative dose- response curves to phenylephrine of aortas from Jak2V617F Jak2V617F HC-EC mice treated with vehicle or hydroxyurea (./ak2i r'171 HC-EC vehicle n=4; and Jak2V6I7F HC-EC HU, n=7) (E), with vehicle or ruxolitinib Uak2} 6 ! 71 HC -EC vehicle, n= 5; and Jak2V617F HC-EC Ruxolitinib n=4) (J), and with vehicle or simvastatin (Jak2V617F HC-EC vehicle, n=10; and Jak2V6I7F HC- EC simvastatin in, n=7) (O). Data are expressed as mean with standard error of the mean for cumulative curve and median with interquartile range for spleen weight and blood cell count. Abbreviations: * p<0.05, ** p< 0.01, HU hydroxyurea; ns, not significant. Cumulative dose response curves were compared using an analysis of variance for repeated measures and other data were compared using the Mann- Whitney U-test. All tests were 2 side
EXAMPLE:
Methods
Experimental design
The objective of our study was to analyse endothelial reactivity in MPN. We first noticed a major increase in arterial contraction in Jak2V617F HC-EC mice, a model with Jak2V617F expression both in hematopoietic and endothelial cells, that mimics the human disease. We then created mouse models specifically mutated in endothelial or in hematopoietic cells. We then searched for the mediators responsible for the increased response to vasoconstrictors when Jak2V6I7F was present in hematopoietic cells and tested the hypothesis that circulating blood might convey biological information from hematopoietic cells to the vascular wall and focused on microvesicles. We identified that erythrocyte-derived microvesicles were responsible for this effect and performed a mass spectrography analysis to highlight the proteins involved. Sample size was chosen based on previous works using the same technique (myography) and microvesicles, published by our team (48, 49).
Mouse breeding occurred in our animal facility in accordance with the local recommendations. Control mice were littermate, appropriate, age, sex and genetic background, matched to account for any variation in data. Institutional animal care and use committee at INSERM (Descartes university, Paris, France) approved all animal experiments (CEEA- 17053).
Number of experimental replicates is provided in each figure legend and included at least 3 independent experiments. For each myography experiment, duplicates with the same aorta were used, averaged and counted as n=l . There was no randomization in these experiments. We did not exclude any other sample than those not fulfilling the quality criteria detailed in the corresponding methods section. Only aorta with a viable endothelium were used for myography (see corresponding methods section for criteria).
For human samples, inclusion and exclusion criteria were defined prior to sample collection (see corresponding methods section for criteria). No outlier was excluded. Investigators were not blinded to group allocation during collection and analysis of the data. All patients (carrying Jak2V617F with a past history of splanchnic vein thrombosis, not receiving any specific treatments other than vitamin-k antagonists) and healthy volunteers gave writing consent to the study. Human study was performed in accordance with the ethical guidelines of the 1975 Declaration of Helsinki and was approved by the institutional review board Bichat- Claude-Bemard (Paris; France).
Murine models
All mice were on a C57BL/6 background. Mice carrying constitutive Jak2l Y'171 mutation in endothelial and hematopoietic cells were obtained by crossing VE-cadherin-Cre transgenic mice provided by M. Souyri (13) with J 2(/577 Flex/WT mice provided by M. Villeval (50). Mice carrying inducible Jak2V617F mutation specifically in endothelial cells were obtained by crossing VE-Cadherin-cre-ERT2 transgenic mice provided by R. Adams (51) with Jak2V6I7F Flex/WT mice provided by M. Villeval (50). The Flex (for Flip-Excision) strategy allows the expression of a mutated gene in adulthood, in a temporal and tissue-specific manner (52). It allows an efficient and reliable Cre-mediated genetic switch: the expression of a given gene is turned on by inversion, while expression of another one is simultaneously turned off by excision. In all experiments, male and female mice were used.
For organ chamber experiments and femoral in vivo experiments, mice were euthanized between the ages of 8 and 17 weeks. For induction of Cre recombinase expression in Jak2V6!7F Flex/WT; VE-Cadherin-cre-ERT2 mice, mice were injected intraperitoneally with tamoxifen (Sigma, T5648), 1 mg/mouse/day for 5 consecutive days over 2 consecutive weeks (10 mg in total per mouse) between the ages of 5 to 7 weeks. Experiments were performed between 4 to 6 weeks after the last tamoxifen injection. Both female and male were used for each experiment.
Experiments were conducted according to the French veterinary guidelines and those formulated by the European community for experimental animal use (L358-86/609EEC) and were approved by the French ministry of agriculture (n° A75-15-32).
Verification of the efficient endothelial recombination in mouse models
All mice were on a C57BL/6 background. Mice with the mTmG reporter provided by C. James (Inserm 1034) were crossed with VE-cadherin-Cre transgenic mice provided by M. Souyri or VE-Cadherin-cre-ERT2 transgenic mice provided by R. Adams (51). For induction of mTmG;VE-Cadherin-cre-ERT2 model, mice were injected intraperitoneally with tamoxifen (Sigma, T5648), 1 mg/mice/day for 5 consecutive days over 2 consecutive weeks (10 mg in total per mice) between the ages of 5 to 7 weeks, and experiments were performed 2 weeks after the last injection of tamoxifen. Aortas and femurs were harvested under isoflurane anaesthesia and fixed in 4% PFA. Aorta were mounted“en face” on glass slides, while femurs were cryosectioned. All tissues were imaged using a Leica SP5 confocal microscope (Leica) at 400 X magnification. For flow cytometry analysis in Cre;mT/mG mice, bone marrow cells were stained with TER - 119 APC and Gr - 1 APC (553673, Becton Dickinson) and analyzed on an
Accuri C6 flow cytometer (BD Biosciences). Data were interpreted using BD Accuri C6 Analysis Software.
Patient's inclusion
All patients fulfilling inclusion criteria were prospectively included at the Hepatology department, Beaujon Hospital, Clichy, France, between May 2016 and July 2016. Only patients carrying Jak2V617F without specific treatment for MPNs were included. All patients had a past history of Budd-Chiari syndrome or portal vein thrombosis and were receiving vitamin K antagonists. Controls were healthy voluntaries. All patients and controls gave written consent to the study. This study was performed in accordance with the ethical guidelines of the 1975 Declaration of Helsinki and was approved by our institutional review board (CPP lie de France IV, Paris; France).
Organ Chamber Experiments
Thoracic aortas from adult mice were isolated after animal sacrifice under 2% isoflurane anaesthesia. The aortic rings were mounted immediately in organ chambers (Multi WireMyograph system, model 610 M; Danish Myo Technology, Aarhus, Denmark) filled with Krebs-Ringer solution (NaCl 118.3 mmol/L, KC1 4.7 mmol/L, MgS04 1.2 mmol/L, KH2P04 1.2 mmol/L, CaC12 1.25 mmol/L, NaHC03 25.0 mmol/L and glucose 5 0 mmol/L) gassed with a mixture of 02 95% and C02 5% (pH 7.4). The presence of functional endothelial cells was confirmed by the relaxation to acetylcholine chloride (Sigma, A6625) (10-5 mol/L) following a contraction evoked by phenylephrine (10-7 mol/L) and was defined as a relaxation > 70% of the precontraction as previously described (49). After extensive washout and equilibration, contraction to phenylephrine hydrochloride (concentration-response curve, 10-9 to 10-4 mol/L) (Sigma, P1250000) or angiotensin II (concentration-response curve, 10-9 to 10-6 mol/L) (Sigma, A9525) or KCL (80 mmol/L) and relaxation to acetylcholine chloride (concentration-response curve, 10-9 to 10-4 mol/L) or SNAP (S-Nitroso-N-acetyl-DL- penicillamine, Sigma, N3398) (concentration-response curve, 10-10 to 10-5 mol/L) were studied. For NO synthase inhibition, aorta rings were preincubated for 45 min with L-NAME 10-4 mol/L (Cayman, 80210) prior to concentration-response curve to phenylephrine without washout. In some experiments, the endothelium was mechanically removed by inserting the tip of a pair of forceps within the lumen and by gently rubbing the ring back and forth on a piece of wet tissue. For the N- Acetyl- Cysteine experiment (NAC, commercial HIDONAC®, Zambon), NAC was added to the Krebs-Ringer solution at a final concentration of 20 mmol/L.
In vivo femoral reactivity
The femoral artery was carefully exposed from adult mice under 2% isoflurane anaesthesia.
Krebs-Ringer solution (cf organ chamber experiment) gassed with a mixture of 02 95% and C02 5% (pH 7.4) at 37° was permanently superfuse (2 mL/min) on the exposed artery. After a 15-minute equilibration, arterial responses were determined by addition of phenylephrine (10-3 mol/L) for 2 min then acetylcholine (10-1 mol/L) for 1 min (Sigma Chemical Co.). On the controlateral leg, with the same protocol, KCL (80 mmol/L) then SNAP (S-Nitroso-N-acetyl-DL-penicillamine, 10-3 mol/L) were used. All dilutions were prepared just before application. Changes in vessel diameter were continuously recorded on a videotape recorder. Subsequently, images were exported and vessel outer diameters were analyzed using Image J Software.
Isolation and characterization of patients’ circulating microvesicles
Circulating microvesicles from patients or healthy control were isolated from platelet- free plasma obtained by successive centrifugations of venous blood, as reported previously (53). Briefly, citrated venous blood (15 mL) was centrifuged twice at 2500g for 15 minutes (at room temperature) to remove cells and cell debris and to obtain platelet-free plasma (PFP). A portion of this PFP was then aliquoted and stored at -80°C. The rest was centrifuged at 20500g for 2 hours (4°C). Supernatant of this 20500g centrifugation was then discarded and the resulting microvesicles pellet was resuspended in a minimal volume of supernatant, aliquoted and stored at -80°C. For each patient, concentrations of annexin V positive microvesicles were analysed in the PFP and the resuspended pellet of microvesicles.
Circulating levels of annexin V+ microvesicles (IM3614, BD) were determined on a Gallios flow cytometer (Beckman Coulter, Villepinte, France) using a technique previously described in detail (49, 53).
Generation of microvesicles from mice
Blood samples were collected from the inferior vena cava of Jak2V617F HC-EC mice or littermate controls using a 25 G x G needle in a 1 mL syringe pre-coated with 3.8% sodium citrate. PFPs were generated as described above for patients and used to measure plasma annexin V positive microvesicles in mice. The pelleted cells obtained following the first 2500g centrifugation were resuspended in PBS to a final volume of 5 mL for control mice and 10 mL
for Jak2V6I7F HC-EC mice. PBMC, PMNC and erythrocytes were separated using a double percoll gradient (63% and 72% for control mice and 63% and 66% for Jak2V l7! HC-EC mice) using a 700g centrifugation for 25 min, without brake. The slight differences between the protocols used for control and Jak2V617F HC-EC mice are the results of the preliminary experiments we did to obtain pure isolation of each cell type. Cells were subsequently washed with PBS, then incubated with 5 gmol/L ionomycin TBS for 30min at 37°C to induce microvesicles generation. 5 mmol/L EDTA was then added to chelate free calcium. Cells were then discarded by centrifugations at 15000g for 1 min and the supernatants were collected. Microvesicles were isolated, as described above using a 20500g centrifugation during 45min. Concentrations of annexin V positive microvesicles (as described above) were analysed in the PFP and the 20500g microvesicles pellet for each mouse.
To isolate platelets, 500 pL of whole blood were diluted in 10 mL of PBS. A 1.063 g/mL density barrier was created by combining 5 mL of 1.320 g/mL 60% iodixanol stock solution (OptiPrep density gradient medium, Sigma-Aldrich, Saint Louis, MO, USA) with 22 mL of diluent (0.85% NaCl, 20 mM HEPES-NaOH, pH 7.4, 1 mM EDTA). For platelet separation, 10 mL of each diluted blood were layered over 10 mL of density barrier and centrifuged at 350 g for 15 minutes at 20°C with the brake turned off. The interface between the density barrier and the blood contained platelets. Residual contaminating erythrocytes were removed by magnetic sorting. Briefly, the cell suspension was labelled with Anti-Ter-119 MicroBeads (Miltenyi Biotec ref 130-049-901) and erythrocytes (Ter- 119+) were negatively sorted using a MACS® Separator. The remaining cells (platelets) were subsequently washed with PBS and exposed to 5 pmol/L ionomycin in TBS for 30 minutes at 37°C. 5 mM EDTA was then added to chelate free calcium. Finally, cells were discarded by centrifugation at 15000g for 1 minute, the supernatant was collected and microvesicles isolated, as previously described.
Vascular reactivity following exposure to microvesicles
For organ chamber experiments, thoracic aortas from adult C57BL/6 mice (8 to 10 weeks old) were isolated after sacrifice under isoflurane anaesthesia. Mouse aortic rings were incubated for 24 hrs; 37°C in a 5% C02 incubator, with filtered DMEM supplemented with antibiotics (100 IU/mL streptomycin, 100 IU/mL penicillin (Gibco, Invitrogen, Paisley, Scotland), and 10 pg/mL polymyxin B (Sigma, St Louis, MO) in the presence of microvesicles. Aortic rings were then mounted in organ chambers and concentration-response curves to pharmacological agents were performed.
For femoral artery in vivo experiment, C57BL/6 mice were injected intravenously (retro-orbital injection) with microvesicles (100 pL final volume with 2 pL heparin sodium (5000 IU/mL)). Experiments were performed 2 hours after injection.
Microvesicles from patients and healthy controls were incubated at their respective individual plasma concentration (Annexin-V positive microvesicles). Microvesicles generated from mice were incubated or injected at the same final concentration for Juki1 6171 HC-EC mice and control mice, namely 7000 Annexin V positive microvesicles / pL for erythrocyte and platelet-derived microvesicles and 700 Annexin V positive microvesicles / pL for PBMC and PMNC-derived microvesicles. We chose these concentrations since we found in preliminary experiments that the majority of mice have concentrations of Annexin V positive microvesicles between 1000 and 10000 / pL, and because PBMC and PMNC-derived microvesicles are consistently found less abundant in the blood than erythrocyte and platelet-derived microvesicles (27, 32).
Bone marrow transplantation
We subjected 6 to 8 weeks old C57B1/6J mice to medullar aplasia following 9.5 gray lethal total body irradiation. We repopulated the mice with an intravenous injection of bone marrow cells isolated from femurs and tibias of age matched Jak2V617F HC-EC and of littermate control mice. Medullar reconstitution was allowed for 8 weeks before experiments.
Treatments
Hydroxyurea (Sigma, H8627), or the same volume of vehicle (NaCl 0.9%), was administrated for 10 consecutive days (100 mg/kg/day BID) by intra-peritoneal injections.
Ruxolitinib (Novartis) was administered for 21 consecutive days (30 mg/kg 2 times per day) by oral gavage (54). Ruxolitinib was prepared from 15-mg commercial tablets in PEG300/5% dextrose mixed at a 1 :3 ratio, as previously reported (55). Control mice were administered the same volume of vehicle (PEG300/dextrose 5%).
Simvastatin (Sigma S6196) was administered for 14 days (20 mg/kg/day, once a day) by intra-peritoneal injections. Activation by hydrolysis was first achieved by dissolving 50 mg in 1 mL of pure ethanol and adding 0.813 ml of 1 mol/L NaOH. pH was adjusted to 7.2 by adding small quantities of 1 mol/L HC1 and dilution was then performed in PBS (56). Control mice were injected with the same volume of vehicle.
Human recombinant Epoietin alfa (5000 UEkg, diluted in 0.2% BSA in PBS) or vehicle (0.2% BSA in PBS) was administered to wild type mice every 2 days for 3 weeks by intraperitoneal injection, as previously described (57).
N-Acetyl-Cysteine (commercial HIDONAC, Zambon) diluted in NaCl 0.9% or the same volume of vehicle (NaCl 0.9%), was administrated for 14 consecutive days (500 mg/kg/day) by intraperitoneal injections.
Blood Cell count analysis
Blood was collected on the day of sacrifice from the inferior vena cava using a 25G x 1’ needle in a 1 mL syringe pre-coated with 3.8% sodium citrate. Blood counts analyses were performed using a Hemavet 950FS analyser (Drew scientific).
Quantification of reactive oxygen species generation
Thoracic aortas from adult mice were isolated after animal sacrifice under 2% isoflurane anaesthesia, longitudinally opened and placed directly in HBSS (Hanks' balanced salt solution, Sigma, 14025-092). For each set of experiments, all aortas were processed immediately after removal, at the same time, with the same reagents and in the same manner. No plasma factor or blood cells were added during the ROS generation assessment. For positive and negative controls, 2 pieces of wild type aortas were incubated with H202 (100 pmol/L final concentration) for 20 min at 37°C. For negative controls, N-Acetyl-Cysteine (5 mmol/L final concentration) was incubated together with H202 for 20 min at 37°C. All aortas were then incubated with 5 pmol/L CellROX® (Fisher scientific, C10422) for 30 min at 37°C. CellROX® Deep Red Reagent is a fluorogenic probe designed to reliably measure reactive oxygen species inside living cells. The cell-permeable CellROX® Deep Red dye is nonfluorescent while outside of the cell and in a reduced state and, upon oxidation, exhibits excitation/emission maxima at 640/665 nm. After rinsing and fixation (Paraformaldehyde 4%, 20 min), samples were costained with DAPI (0.1 pg mL, Sigma) in order to identify cell nuclei. After staining, aortas were washed with PBS, mounted“en face” on glass slides and imaged using a bright field Zeiss Axio Imager Z1 (Zeiss) microscope. Images were acquired in the 2 hours following staining at 400 X magnification. CellROX® positive surface (in red) and the number of cells were quantified using Image J Software.
Myeloperoxidase inhibition in microvesicles
Erythrocyte-derived microvesicles from Jak2V617F HC-EC mice were incubated for 1 hr with an irreversible MPO inhibitor (MPOi, PF06281355, resuspended in DMSO, Sigma), diluted in PBS (5 Dmol/L final concentration). Then, the same amount of annexin V positive erythrocyte-derived microvesicles (JAK2WT, Jak2V617F and Jak2V617F with MPOi) were washed in PBS and centrifuged at 20500g for 2 hrs. The pellet containing the microvesicles was then resuspended in endothelial cell basic medium (Promocell). HUVECs (single donor, C-12200, lot 445Z011, PromoCell) were then incubated for 2 hrs at 37°C with these microvesicles. At the end of the incubation, and without washing cells, reactive oxygen species generation was assessed using CELLROX® (Fisher scientific, Cl 0422), as described above. After rinsing with medium and paraformaldehyde (4%, 5 min), HUVECs were costained with DAPI (0.1 pg/mL, Sigma) in order to identify nuclei. Images were acquired using a Confocal microscope, Leica SP8 at 400 X magnification.
Electrocardiography
Electrocardiograms were recorded from mice using the non-invasive ecgTUNNEL (Emka Technologies) with minimal filtering Electrocardiogram was continuously monitored for 3 min (baseline). Waveforms were recorded using Iox Software and heart rate and intervals were measured with ECG Auto from recording traces. Following baseline determination, the animals received a single administration of phenylephrine (bolus, 3mg/Kg) by intravenous route at the caudal vein and electrocardiogram were recording 3-5 minutes more.
RNA gene allelic discrimination
Erythrocytes microvesicles were lysed with Qiazol lysis reagent (Qiagen) and RNA was extracted with RNeasy micro Kit (Qiagen) according to manufacturer’s instructions. Dosage of RNA was performed with Qubit HS RNA assay Kit (ThermoFisher Scientitic). cDNA synthesis was performed with QuantiTect Reverse Transcription Kit (Qiagen). RNA gene allelic discrimination was performed by Taqman analysis with the ABI Prism GeneAmp 7500 Sequence Detection System (Applied Biosystem, Invitrogen) using as primers: TTTACAAATTCTTGAACCAGAATGTTC (JAK2 forward - SEQ ID NO: 1) and TTCTCACAAGCATTTGGTTTTGAAT (JAK2 reverse SEQ ID NO: 2) and as probes: VIC- CTCCACAGACACAGAC-MGB for JAK2WT (SEQ ID NO: 3) and 6-FAM- TCTCCACAGAAACAGAGA-MGB for Jak2Vf'm (SEQ ID NO: 4)
Mass spectrometry analysis
Size-exclusion chromatography of microvesicles was then performed in order to separate microvesicles from soluble proteins. Successive aliquot of 150 pL were collected and measurement of protein absorbance was performed. Fractions contained in tubes 6 to 11, containing microvesicles, were selected and then centrifuged at 20 500g for two hours. To finish, microvesicles were lysed using a 1% triton buffer.
For mass spectrometry analysis, proteins were precipitated overnight at -20°C with 0.1 mol/L Ammonium Acetate glacial in 80% methanol (buffer 1). After centrifugation at 14000xg and 4°C for 15 min, the resulting pellets were washed twice with 100 pL of buffer 1 and further dried under vacuum (Speed-Vac concentrator). Proteins were then reduced by incubation with 10 pL of 5 mmol/L dithiotreitol (DTT) at 57°C for one hour and alkylated with 2 pL of 55 mmol/L iodoacetamide for 30 min at room temperature in the dark. Trypsin/LysC (Promega) was added twice at 1 : 100 (wt:wt) enzyme: substrate, at 37°C for 2 hrs first and then overnight. Samples were then loaded onto a homemade Cl 8 StageTips for desalting. Peptides were eluted using 40/60 MeCN/H20 + 0.1% formic acid and vacuum concentrated to dryness. Online chromatography was performed with an RSLCnano system (Ultimate 3000, Thermo Scientific) coupled online to a Q Exactive F1F-X with a Nanospay Flex ion source (Thermo Scientific) Peptides were first trapped on a Cl 8 column (75 pm inner diameter x 2 cm; nanoViper Acclaim PepMapTM 100, Thermo Scientific) with buffer A (2/98 MeCN/H20 in 0.1% formic acid) at a flow rate of 2.5 pL/min over 4 min. Separation was then performed on a 50 cm x 75 pm Cl 8 column (nanoViper Acclaim PepMapTM RSLC, 2 pm, lOOA, Thermo Scientific) regulated to a temperature of 50°C with a linear gradient of 2% to 30% buffer B (100% MeCN in 0.1% formic acid) at a flow rate of 300 nL/min over 91 min. MS full scans were performed in the ultrahigh-field Orbitrap mass analyzer in ranges m/z 375-1500 with a resolution of 120 000 at m/z 200. The top 20 intense ions were subjected to Orbitrap for further fragmentation via high energy collision dissociation (HCD) activation and a resolution of 15 000 with the intensity threshold kept at 1.3 x 105. We selected ions with charge state from 2+ to 6+ for screening. Normalized collision energy (NCE) was set at 27 and the dynamic exclusion of 40s.
For identification, data were searched against the Mus Musculus one gene one protein (UP000000589_10090) UniProt database and a databank of the common contaminants using Sequest F1F through proteome discoverer (version 2.2). Enzyme specificity was set to trypsin and a maximum of two-missed cleavage sites were allowed. Oxidized methionine and N- terminal acetylation were set as variable modifications. Maximum allowed mass deviation was set to 10 ppm for monoisotopic precursor ions and 0.02 Da for MS MS peaks. The resulting files were further processed using myProMS (58) v3.6. FDR calculation used Percolator and
was set to 1% at the peptide level for the whole study. The label free quantification was performed by peptide Extracted Ion Chromatograms (XICs) computed with MassChroQ version 2.2 (59). For protein quantification, XICs from proteotypic peptides shared between compared conditions (TopN matching) with two-missed cleavages were used. Median and scale normalization was applied on the total signal to correct the XICs for each biological replicate. To estimate the significance of the change in protein abundance, a linear model (adjusted on peptides and biological replicates) was performed and p-values were adjusted with a Benjamini-Hochberg FDR procedure with a control threshold set to 0.05.
The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE (60) partner repository with the dataset identifier PXD014451.
Western Blot on erythrocyte-derived microvesicles
Erythrocyte-derived microvesicles generated as mentioned above were centrifuged at 20 500g for 2 hrs, and then lysed in 100 DL RIPA buffer containing 150 mmol/L NaCl, 50 mmol/L TrisHCl, pH 7.4, 2 mmol/L EDTA, 0.5% sodium deoxycholate, 0.2% sodium dodecyl sulfate, 2 mmol/L activated orthovanadate, complete protease inhibitor cocktail tablet (Complet mini, Roche, France) and complete phosphatase inhibitor cocktail tablet (Roche, France). Protein content was quantified using the Micro BCA Protein Assay Kit (Thermo Scientific). Equal loading was checked using Ponceau red solution. Membranes were incubated with primary antibodies (1/1000) (Anti-GP91, BD611415; Anti-GSTTl, Abcaml99337; AntiMPO, Abcam45977). After secondary antibody incubation (anti-rat, Cell Signaling, 1/1000; anti rabbit or anti-mouse, Amersham, GE Healthcare, UK 1/3000), immunodetection was performed using an enhanced chemiluminescence kit (Immun-Star Western C kit, Bio-Rad). Bands were revealed using the LAS-4000 imaging system. Values reported from Western blots were obtained by band density analysis using with ImageJ software and expressed as the ratio protein of interest compared to Ponceau.
Uptake of microvesicles by endothelial cells
Erythrocyte-derived microvesicles were stained with PKH26 dye (Sigma Aldrich) diluted in PBS, following manufacturer’s instructions, washed in PBS and then centrifuged at 20500g for 2 hrs. The 20500g supernatant served for control experiments. Murine endothelial cells (the cell line called SVEC4-10, CRL-2181, lot 70008729, ATCC) were then incubated with these stained microvesicles or an equal volume of the 20500g supernatant. After 2 hrs at 37°C, cells were washed 3 times with DMEM (Gibco). Cells were then fixed in PFA 4% for 5
min, and then costained with DAPI (0.1 Dg/mL, Sigma) in order to identify cell nuclei. Images were acquired using a Confocal microscope, Leica SP8 at 600 X magnification.
Statistics
For cumulative dose response curves, data were expressed as mean with standard error of the mean and compared using an analysis of variance for repeated measures. Other data were expressed as median with interquartile range (blood cell count and spleen weight) and compared using the Mann- Whitney U-test. All tests were 2 sided and used a significance level of 0.05. Data handling and analysis were performed with GraphPad Software, Inc.
Study approval
Institutional animal care and use committee at INSERM (Descartes university, Paris, France) approved all animal experiments (CEEA- 17-053).
For human samples, inclusion and exclusion criteria were defined prior to sample collection (see corresponding methods section for criteria). All patients (carrying Jak2V617F with a past history of splanchnic vein thrombosis, not receiving any specific treatments other than vitamin-k antagonists) and healthy volunteers gave writing consent to the study. Human study was performed in accordance with the ethical guidelines of the 1975 Declaration of Helsinki and was approved by the institutional review board (CPP He de France IV, Paris; France).
Results
Increased arterial contraction in mice carrying Jak2V6I7F in hematopoietic and endothelial cells
As Jak2V617Fis present in both hematopoietic and endothelial cells in patients with MPN (2, 3), we first investigated vasoactive response in a mouse model mimicking the human disease. We generated mice expressing Jak2V617F in hematopoietic and in endothelial cells by crossing Jak2V6I7F Flex/WT mice with VE-Cadherin-cre mice. VE-Cadherin being expressed during early embryonic life in a precursor of both endothelial and hematopoietic cells (13), Jak2V6I7F Flex/WT; VE-Cadherin-cre, thereafter referred to as Jak2V6I7F HC-EC, developed as expected a MPN, attested by higher spleen weight (2.3 to 5.7 % of body weight vs. 0.3 to 0.6 % for littermate controls, pO.OOOl), and higher haemoglobin level, platelet and white blood cell counts than littermate controls (Figures 1A-D). The endothelial and hematopoietic progenitor cells recombination was verified by crossing VE-Cadherin-cre with mTmG mice.
By performing myography assay, we observed ex vivo that aortas from Jak2V617F HC- EC mice displayed a major increase in the response to phenylephrine (Figure IE), but also to potassium chloride (Figure IF) and to angiotensin II (Figure 1G), as compared with littermate controls. Removing the endothelium suppressed this increased arterial contraction (Figure 1H). Likewise, we observed in vivo that femoral arteries from Jak2V617F HC-EC mice displayed an increased response to phenylephrine as compared with littermate controls (Figure II). Thus, Jak2V617F in endothelial and hematopoietic cells strongly increases arterial response to vasoconstrictors in an endothelium-dependent manner.
Because of the high rate of myocardial infarction without significant coronary stenosis reported in patients with MPN (8), we investigated cardiac vascular bed by performing electrocardiography in Jak2V6!7F HC-EC mice and their littermate controls. After intravenous injection of phenylephrine Jak2V617F HC-EC mice displayed electrocardiogram modifications including bradycardia and arrhythmia, that are indirect signs of coronary spasm (14) (Figures 1J-K)
Increased arterial contraction in mice with Jak2V617F specifically expressed in hematopoietic cells but not in endothelial cells
To determine if this increased arterial contraction was due to Jak2V617F in endothelial cells or in hematopoietic cells, we first generated mice expressing Jak2V6I7F only in endothelial cells. We crossed Jak2V6I7F Flex/WT mice with inducible VE-Cadherin-cre-ERT2 mice expressing the ere recombinase after tamoxifen injection only in endothelial cells. As expected, Jak2V6I7F Flex/WT; VE-Cadherin-cre-ERT2 (thereafter referred to as ./ak2i 6l /' EC) mice did not develop MPN (Figures 2A-D). Adequate endothelial recombination was verified by crossing VE-Cadherin-cre-ERT2 mice with mTmG mice. We previously demonstrated the absence of hematopoietic recombination in this model (15). We observed no difference in arterial response to phenylephrine between the Jak2V617F EC mice and littermate controls (Jak2WT) (Figure 2E).
To assess the implication of Jak2V617F in hematopoietic cells, we generated mice expressing Jak2V617F only in hematopoietic cells, by transplanting lethally irradiated C57BL/6 mice with Jak2V617F bone marrow cells obtained from Jak2V617F HC-EC mice. Irradiated C56BL/6 mice transplanted with Jak2WT BM were used as controls. Hematopoietic expression of Jak2V617F induced a MPN (Figures 2F-I) and an increased arterial response to phenylephrine (Figure 2J). Taken altogether, these findings demonstrate that presence of the Jak2V617F mutation in hematopoietic, but not in endothelial cells, is responsible for the increase in arterial
contraction in response to vasoconstrictors we observed in Jak2V617F HC-EC mice (Figures 1E- G)
Increased arterial contraction induced by microvesicles from Jak2V6I7F patients
We then sought to identify the mediators responsible for the increased response to vasoconstrictors when Jak2V617F was present in hematopoietic cells and tested the hypothesis that circulating blood might convey biological information from hematopoietic cells to the vascular wall. Circulating microvesicles, i.e. extracellular vesicles having a size ranging from 0.1 to 1 pm, are now recognized as triggers of various types of vascular dysfunction (16). We therefore examined the effect of circulating microvesicles isolated from the blood of patients with MPN on vascular responses to vasoactive agents. We isolated plasma microvesicles from 7 patients carrying Jak2V617F (2 males, 5 females; blood drawn before introduction of cytoreductive therapy), and from 5 healthy controls (2 males, 3 females; age not significantly different from patients). We incubated these microvesicles at their plasma concentration with aortic rings from wild type mice and observed that plasma microvesicles from patients carrying Jak2V617F reproduced the increased response to phenylephrine (Figure 3A). Plasma without microvesicles from the same patients and controls had no effect (data not shown).
Increased arterial contraction induced by erythrocytes-derived microvesicles from Jak2V617F mice
We then sought to determine the subpopulation of microvesicles responsible for this increased arterial contraction. We generated microvesicles from each type of blood cells from Jak2V617F HC-EC mice or littermate controls and incubated these microvesicles, at the same concentration, with aortic rings from wild type mice. Erythrocyte-derived microvesicles generated from Jak2V617F HC-EC mice reproduced the increased response to phenylephrine on aortic rings ex vivo (Figure 3E), while platelet, peripheral blood mononuclear cell and polynuclear neutrophil microvesicles did not (Figures 3B-D). Likewise, in vivo, femoral arteries from wild-type mice injected with microvesicles derived from Jak2V617F erythrocytes displayed an increased response to phenylephrine as compared with wild-type mice injected with microvesicles derived from littermate controls’ erythrocytes (Figure 3F). Microvesicles generated from Jak2V617F HC-EC mice erythrocytes carried Jak2V617F mRNA (Figure 3G). To investigate the interaction of erythrocyte-derived microvesicles with endothelial cells, we labeled microvesicles with the fluorescent dye PKH-26, incubated them with endothelial cells and then performed confocal microscopy on endothelial cells. Fluorescence was detected in endothelial cells, suggesting that erythrocyte-derived microvesicles were taken-up by
endothelial cells (17). No difference in uptake was observed between microvesicles from Jak2V617F HC-EC erythrocytes and their wild-type counterparts (Figure 3H).
We then wanted to determine whether the increased number of erythrocytes could in itself explain this effect or if qualitative changes were involved. We generated a mouse model of polycythaemia without Jak2V6!7F , caused by chronic epoietin injections. After 3 weeks of epoietin treatment, haemoglobin reached a level similar to that of Jak2V6I7F HC-EC mice (18.5 g/dL, interquartile range 16.5-19.5, vs. 17.6 g/dL, interquartile range 15.7-19.7, respectively; n=5 and n=13 respectively; p=0.67). However, this model with high number of circulating erythrocytes failed to reproduce the increased response to phenylephrine observed in Jak2V6,7F HC-EC mice aortas (Figure 31). Thus, the presence of the Jak2V617F mutation in erythrocyte- derived microvesicles is required to cause increased arterial contraction.
NO pathway inhibition and endothelial increased oxidative stress status
We then investigated how microvesicles derived from Jak2V617F erythrocytes increase response to vasoconstrictive agents.
We examined first the nitric oxide (NO) pathway. We observed ex vivo on aortas and in vivo on femoral arteries that Jak2V617F HC-EC mice, reproducing the human disease, display an impaired dilatation to acetylcholine (Figures 4A, B). This impaired dilatation capacity was not due to decreased sensitivity to NO of vascular smooth muscle cells, as the response to a direct NO-donor (SNAP) was not different between Jak2V617F HC-EC mice and littermate controls, both ex vivo and in vivo (Figures 4C, D). We also observed that, after pre-incubation with the NO synthase (NOS) inhibitor L-Name, aorta from Jak2V617F HC-EC mice had a similar response to phenylephrine as littermate controls (Figure 4E). Therefore, these results demonstrate that the increased arterial contraction observed in Jak2V617F HC-EC mice results from a dysfunctional endothelial NO pathway.
Because previous works showed that heme in erythrocytes microvesicles can scavenge NO (18, 19), we quantified heme in microvesicles derived from Jak2V617F HC-EC mice and control mice erythrocytes, but observed no difference.
We then investigated generation of reactive oxygen species, i.e. inhibitors of NOS activity and NO bioavailability (20) and demonstrated 4 times more reactive oxygen species in the aortic endothelium of Jak2V6I7F HC-EC mice than in littermate controls (Figure 4F). Conversely, reactive oxygen species generation was normal in the aortic endothelium of Jak2V6I7F EC mice, expressi ng Jak21 7 ' 7 only in endothelial cells (Figure 4G). Likewise, aortic endothelium from wild-type mice injected with microvesicles derived from Jak2V6I7F
erythrocytes displayed more reactive species generation than aortic endothelium of mice injected with microvesicles derived from littermate controls erythrocytes (Figure 4H). There was no reactive oxygen species generation in underlying smooth muscle cells in any of these experiments (data not shown). Altogether, these results show that microvesicles derived from JAK2V617F erythrocytes induce an excessive oxidative stress in endothelial cells leading to a decreased availability of NO.
To ascertain the implication of the increased oxidative stress in the increased arterial contraction in Jak2V617F HC-EC mice, we treated these mice with N-Acetyl-Cysteine (NAC, activator of glutathione pathway and anti-oxidant) for 14 days intraperitoneally. This treatment had no effect on blood cell count or spleen weight but normalized arterial contraction to phenylephrine (Figure 41).
Increased endothelial oxidative stress status by myeloperoxidase in erythrocyte- derived microvesicles from Jak2V617F mice
To shed light on the mechanisms underlying this increased oxidative stress induced by microvesicles derived from Jak2V6I7F erythrocytes, we performed a proteomic analysis of these microvesicles (data are available via ProteomeXchange with identifier PXD014451). All proteins involved in reactive oxygen species detoxification or generation are shown in the volcano-plot shown in Figure 5A. We found one protein significantly deregulated (glutathione S transferase theta 1, GSTT1) and one protein only detected in microvesicles derived from Jak2V6I7F erythrocytes (myeloperoxidase, MPO) that could explain the observed effect (Figure 5A). We also considered cytochrome b-245 heavy and light chain (NOX2) although only few peptides were detected in microvesicles derived from Jak2V617F erythrocytes (1 peptide of heavy chain in 1/6 samples and 1 peptide of light chain in 2/6 samples), since NOX plays a key role in oxidative stress and since no NOX2 peptide was detected in microvesicles derived from JAK2WT erythrocytes. Western blot analyses were then performed on microvesicles derived from Jak2V617F and control erythrocytes to verify proteomic results. By western blot, NOX2 expression evaluated by Gp91 was not significantly different between Jak2V6I7F and controls erythrocyte-derived microvesicles. GSTT1, which has an anti-oxidant effect (21), was significantly lower in Jak2V617F than in control erythrocyte-derived microvesicles (Figure 5B). Expression of MPO, a protein with a strong pro-oxidant effect (22) was much higher in Jak2V617F than in control erythrocyte-derived microvesicles (Figure 5C). We then directly inhibited MPO in microvesicles derived from erythrocytes of Jak2V617F HC-EC mice before incubation with endothelial cells (HUVEC) in vitro. We observed that the MPO inhibitor
(PF06281355) completely reversed the increase in endothelial oxidative stress induced by Jak2V617F erythrocyte-derived microvesicles (Figure 5D).
In conclusion, Jak2V6I7F erythrocyte-derived microvesicles carry MPO that confers a pro-oxidant phenotype to endothelial cells, leading to increased arterial contraction observed in Jak2V6]7F ¥LC-EC mice.
Statins as a potential new treatment in myeloproliferative neoplasms
We then tested if available treatments for MPNs, namely hydroxyurea and ruxolitinib, affect this increased arterial contraction. ln Jak2V617FY£-EC mice, best representing the human disease, hydroxyurea for 10 consecutive days decreased spleen weight, haemoglobin level and white blood cell count (Figures 6A, 6B, 6D). However, platelet count was not affected by this short time hydroxyurea treatment (Figure 6C). Hydroxyurea significantly improved contraction in response to phenylephrine as compared to vehicle (Figure 6E).
We then treated Jak2V617F HC-EC mice with ruxolitinib for 21 consecutive days and observed a significant decrease in the spleen weight and white blood cell count (Figures 6F, 61), but no effect on the haemoglobin level and on platelet count (Figures 6G, 6H) Ruxolitinib had no effect on arterial response to phenylephrine (Figure 6J).
Beyond its lowering cholesterol effect, simvastatin also improves endothelial function through NO pathway and by preventing oxidative stress damage (23, 24). Thus, we tested its effect on arterial response to phenylephrine in Jak2V617F HC-EC mice. Fourteen days of treatment with simvastatin did not change spleen weight, haemoglobin level or platelet count (Figures 6 K-M). There was only a slight decrease in white blood cells count following simvastatin treatment (Figure 6N). Interestingly, simvastatin significantly improved aortic response to phenylephrine as compared to vehicle (Figure 60).
Discussion:
This study demonstrated that Jak2V617F erythrocyte-derived microvesicles carrying MPO, are responsible for an increased oxidative stress in arterial endothelium and a decreased availability of NO, which strongly increased arterial contraction to vasoconstrictive agents, possibly accounting for arterial events associated with MPNs. Simvastatin, a drug with anti oxidant properties, improved arterial contraction.
The first major finding of our study is the demonstration that Jak2V617F MPN induces a considerable increase in arterial contraction. This finding suggests a vasospastic phenomenon associated with MPN and thus represents a paradigm shift in MPNs where arterial events were
only seen as a result of a thrombotic process (7). Our results obtained ex vivo and in vivo could explain this higher incidence of arterial events in patients with polycythemia vera than in the general population and the high prevalence of myocardial infarction without significant coronary stenosis by angiography in patients with MPN (8). Arterial spasm is an underdiagnosed phenomenon that can happen in patients without atherosclerosis but is also favoured by underlying non-stenotic atherosclerotic plaques. This suggests that the effect we observed might not only account for myocardial infarction without significant coronary stenosis reported in patients with MPN, but might also more widely favour arterial events in patients with atherosclerotic plaques and MPN (11, 12). Moreover, arterial spasm not only occurs in coronary arteries, but also in brain arteries (25). We also found an impairment in arterial dilatation, which is in line with the altered endothelial dependant flow mediated vasodilatation reported in patients with polycythemia vera, in the absence of overt arterial disease (26).
The second major finding of our work is the contribution of Jak2V617F erythrocyte- derived microvesicles to this increased arterial contraction associated with MPN. Importantly, we observed this effect with Jak2V617F erythrocyte microvesicles from mice, but also with microvesicles isolated from patients carrying Jak2V6I7F. We thus highlight here a crucial vascular role of microvesicles in MPNs, beyond their so far described implication in coagulation in this setting (27-31). Although patients with MPNs have higher circulating levels of microvesicles than healthy individuals, we assessed vascular reactivity using the same concentrations of microvesicles for both groups, suggesting that microvesicles composition, and not concentration, accounts for the observed vascular effect (27, 28, 32-35). We cannot rule out the fact that Jak2V6I7F erythrocytes themselves, in addition to microvesicles, could also directly increase arterial contraction associated with MPNs.
Finally, in our work we demonstrated that NO pathway inhibition and increased endothelial oxidative stress are implicated in this increased arterial contraction in MPN. Several groups reported high levels of circulating reactive oxygen species products (36-38) and low antioxidant status in MPN (37, 39), but endothelial oxidative stress had never been investigated. Erythrocyte microvesicles have already been linked to vascular dysfunction in different settings, such as erythrocytes storage or sickle cell disease (18, 19, 40), but never in the context of MPN. Thanks to proteomics approaches we were able to identify a defect in GSTT1 and an over expression of MPO in microvesicles derived from Jak2V617F erythrocytes. Using a direct and irreversible inhibition of MPO, we were able to ascertain the role of MPO carried by microvesicles derived from Jak2V617F erythrocytes in the increase endothelial oxidative stress. MPO is a polycationic heme-containing glycoprotein stored mainly in the azurophilic granules
of neutrophils, but up to 30% of total cellular MPO can be released as active enzyme into the extracellular space. Interestingly, extra-cellular MPO can bind to red blood cells membrane and favours endothelial dysfunction in the context of ischemic heart disease (41-45). Our results demonstrate that MPO binds to erythrocyte-derived microvesicles, increases endothelial oxidative stress and vascular response to vasoconstrictors.
The role of GSTT1 in the increased endothelial oxidative stress status and vascular reactivity we observed with microvesicles derived from Jak2V617F erythrocytes remains uncertain, because we could not restore a normal level of GSTT1 only in microvesicles. The normalisation of vascular reactivity induced by NAC could be explained by the glutathione inducer activity of NAC, but could also just be due to the potent anti-oxidant activity of this drug.
In addition to demonstrating how Jak2V617F induces this increased arterial contraction, our results open new potential therapeutic perspectives to prevent cardio-vascular events in patients with MPN. We demonstrated that simvastatin, a well-known and easily accessible drug, strongly improves arterial response to vasoconstrictive agent in our MPN mouse model. These results thus pave the way for testing simvastatin to prevent arterial events in patients with MPN We also tested available treatments for MPN and observed that hydroxyurea, but not ruxolitinib, improved arterial contraction. This difference might be explained by the fact that hydroxyurea decreased erythrocyte count in our mouse model whereas ruxolitinib did not (46, 47).
In conclusion, our study showed that microvesicles derived from erythrocytes are responsible for an increased arterial contraction in Jak2V617F MPNs. This effect is due to an overexpression of MPO in Jak2V6I7F erythrocyte-derived microvesicles, which is responsible for an increased endothelial oxidative stress and a NO pathway inhibition. Simvastatin appears as an original new approach to prevent arterial events in MPN and warrants further studies.
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. Vainchenker W, Kralovics R. Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood 2017; 129(6):667-679.
2. Sozer S et al. The presence of JAK2V617F mutation in the liver endothelial cells of patients with Budd-Chiari syndrome. Blood 2009;113(21):5246-5249.
3. Rosti V et al. Spleen endothelial cells from patients with myelofibrosis harbor the JAK2V617F mutation. Blood 2013;121(2):360-368.
4. Rosti V et al. High Frequency of Endothelial Colony Forming Cells Marks a Non- Active Myeloproliferative Neoplasm with High Risk of Splanchnic Vein Thrombosis. PLOS ONE 2010;5(12):el5277.
5. Helman Ricardo et al. Granulocyte whole exome sequencing and endothelial JAK2V617F in patients with JAK2V617F positive Budd - Chian Syndrome without myeloproliferative neoplasm. British Journal of Haematology 2018;180(3):443-445.
6. Teofili L et al. Endothelial progenitor cells are clonal and exhibit the JAK2(V617F) mutation in a subset of thrombotic patients with Ph-negative myeloproliferative neoplasms. Blood 2011 ; 1 17(9):2700-2707.
7. Barbui T, Finazzi G, Falanga A. Myeloproliferative neoplasms and thrombosis. Blood 2013; 122(13):2176-2184.
8. Posfai E, Marton I, Borbenyi Z, Nemes A. Myocardial infarction as a thrombotic complication of essential thrombocythemia and polycythemia vera. Anatol J Cardiol 2016; 16(6):397-402.
9. Larsen Al et al. Characteristics and outcomes of patients with acute myocardial infarction and angiographically normal coronary arteries. The American Journal of Cardiology 2005;95(2):261-263.
10. Agewall S et al. ESC working group position paper on myocardial infarction with non-obstructive coronary arteries. Eur Heart J 2017;38(3): 143—153.
11. Davies MJ. The pathophysiology of acute coronary syndromes. Heart 2000;83(3):361-366.
12. Crea F, Libby P. Acute Coronary Syndromes: The Way Forward From Mechanisms to Precision Treatment. Circulation 2017, 136(12): 1155—1166.
13. Oberlin E, El Hafny B, Petit-Cocault L, Souyri M. Definitive human and mouse hematopoiesis originates from the embryonic endothelium: a new class of HSCs based on VE- cadherin expression Int J. Dev. Biol. 2010;54(6-7): 1165-1173.
14. Scherlag BJ, Kabell G, Harrison L, Lazzara R. Mechanisms of bradycardia-induced ventricular arrhythmias in myocardial ischemia and infarction. Circulation 1982;65(7): 1429- 1434.
15. Kilani B et al. Comparison of endothelial promoter efficiency and specificity in mice reveals a subset of Pdgfb-positive hematopoietic cells. J. Thromb. Haemost. 2019;17(5):827- 840.
16. Boulanger CM, Loyer X, Rautou P-E, Amabile N. Extracellular vesicles in coronary artery disease. Nat Rev Cardiol 2017; 14(5):259-272.
17. van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018; 19(4):213-228.
18. Donadee C et al. Nitric oxide scavenging by red blood cell microparticles and cell- free hemoglobin as a mechanism for the red cell storage lesion. Circulation 2011;124(4):465- 476.
19. Camus SM et al. Circulating cell membrane microparticles transfer heme to endothelial cells and trigger vasoocclusions in sickle cell disease. Blood 2015; 125(24):3805- 3814.
20. Yanhoutte PM, Zhao Y, Xu A, Leung SWS. Thirty Years of Saying NO: Sources, Fate, Actions, and Misfortunes of the Endothelium-Derived Vasodilator Mediator. Circ. Res. 2016;119(2):375-396.
21. Stocker R, Keaney JF. Role of Oxidative Modifications in Atherosclerosis. Physiological Reviews 2004;84(4): 1381-1478.
22 Teng N et al. The roles of myeloperoxidase in coronary artery disease and its potential implication in plaque rupture. Redox Report 2017;22(2):51-73.
23. Sen-Baneijee S et al. Kruppel-Like Factor 2 as a Novel Mediator of Statin Effects in Endothelial Cells. Circulation 2005; 112(5):720-726.
24. Mason RP, Walter MF, Jacob RF. Effects of HMG-CoA reductase inhibitors on endothelial function: role of microdomains and oxidative stress. Circulation 2004; 109(21 Suppl 1):II34-41.
25. Bang OY, Toyoda K, Arenillas JF, Liu L, Kim JS. Intracranial Large Artery Disease of Non-Atherosclerotic Origin: Recent Progress and Clinical Implications. J Stroke 2018;20(2):208-217.
26. Neunteufl T, Heher S, Stefenelli T, Pabinger I, Gisslinger H. Endothelial dysfunction in patients with polycythaemia vera. Br. J. Haematol. 2001;115(2):354-359.
27. Charpentier A et al. Microparticle phenotypes are associated with driver mutations and distinct thrombotic risks in essential thrombocythemia. Haematologica 2016;101(9):e365- 368.
28. Tan X et al. Role of erythrocytes and platelets in the hypercoagulable status in polycythemia vera through phosphatidylserine exposure and microparticle generation. Thrombosis and Haemostasis 2013; 109(06): 1025-1032.
29. Baccouche H et al. The evaluation of the relevance of thrombin generation and procoagulant activity in thrombotic risk assessment in BCR-ABL-negative myeloproliferative neoplasm patients. Int J Lab Hematol 2017;39(5):502-507.
30. Duchemin J et al. Increased circulating procoagulant activity and thrombin generation in patients with myeloproliferative neoplasms. Thromb. Res. 2010; 126(3):238-242.
31. Marchetti M et al. Phospholipid-dependent procoagulant activity is highly expressed by circulating microparticles in patients with essential thrombocythemia. Am. J. Hematol. 2014;89(l):68-73.
32. Trappenburg MC et al. Elevated procoagulant microparticles expressing endothelial and platelet markers in essential thrombocythemia. Haematologica 2009;94(7):911-918.
33. Zhang W et al. Clinical significance of circulating microparticles in Ph- myeloproliferative neoplasms. Oncol Lett 2017;14(2):2531-2536.
34 Moles-Moreau M-P et al Flow cytometry-evaluated platelet CD36 expression, reticulated platelets and platelet microparticles in essential thrombocythaemia and secondary thrombocytosis. Thromb. Res. 2010; 126(5):e394-396.
35 Kissova J, Ovesna P, Bulikova A, Zavfelova J, Penka M. Increasing procoagulant activity of circulating microparticles in patients with Philadelphia-negative myeloproliferative neoplasms: a single-centre experience. Blood Coagul. Fibrinolysis 2015;26(4):448-453.
36. Musolino C et al. Changes in advanced oxidation protein products, advanced glycation end products, and s-nitrosylated proteins, in patients affected by polycythemia vera and essential thrombocythemia. Clin. Biochem. 2012;45(16-17): 1439-1443.
37. Durmus A et al. Increased oxidative stress in patients with essential thrombocythemia. Eur Rev Med Pharmacol Sci 2013; 17(21):2860-2866.
38. Vener C et al. Oxidative stress is increased in primary and post-polycythemia vera myelofibrosis. Exp. Hematol. 2010;38(11): 1058-1065.
39. Hasselbalch HC et al. Whole blood transcriptional profiling reveals deregulation of oxidative and antioxidative defence genes in myelofibrosis and related neoplasms. Potential implications of downregulation of Nrf2 for genomic instability and disease progression. PLoS ONE 2014;9(l l):el 12786.
40. Said AS, Doctor A. Influence of red blood cell-derived microparticles upon vasoregulation. Blood Transfus 2017; 15(6):522-534.
41. Adam M et al. Red blood cells serve as intravascular carriers of myeloperoxidase. Journal of Molecular and Cellular Cardiology 2014;74:353-363.
42. Gorudko IV et al. Binding of human myeloperoxidase to red blood cells: Molecular targets and biophysical consequences at the plasma membrane level. Archives of Biochemistry and Biophysics 2016;591 :87-97.
43. Cheng David et al. Inhibition of MPO (Myeloperoxidase) Attenuates Endothelial Dysfunction in Mouse Models of Vascular Inflammation and Atherosclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology 0(0):ATVBAHA.119.312725.
44. Benson TW et al. A single high-fat meal provokes pathological erythrocyte remodeling and increases myeloperoxidase levels: implications for acute coronary syndrome. Laboratory Investigation 2018;98(10): 1300.
45. Baldus S et al. Myeloperoxidase enhances nitric oxide catabolism during myocardial ischemia and reperfusion. Free Radical Biology and Medicine 2004;37(6):902-911.
46. Angona A et al. Dynamics of JAK2 V617F allele burden of CD34+ haematopoietic progenitor cells in patients treated with ruxolitinib. Br. J. Haematol. 2016;172(4):639-642.
47 Vainchenker W et al. JAK inhibitors for the treatment of myeloproliferative neoplasms and other disorders. FlOOORes 2018;7:82.
48. Boulanger CM et al. Circulating Microparticles From Patients With Myocardial Infarction Cause Endothelial Dysfunction. Circulation 2001; 104(22):2649-2652.
49. Rautou P-E et al. Abnormal plasma microparticles impair vasoconstrictor responses in patients with cirrhosis. Gastroenterology 2012; 143(1): 166-176. e6.
50. Marty C et al. A role for reactive oxygen species in JAK2V617F myeloproliferative neoplasm progression. Leukemia 2013;27(11):2187-2195.
51. Wang Y et al. Ephrin-B2 controls VEGF-induced angiogenesis and lymphangiogenesis. Nature 2010;465(7297):483-486.
52. Schniitgen F, Ghyselinck NB. Adopting the good reFLEXes when generating conditional alterations in the mouse genome. Transgenic Res. 2007; 16(4):405-413.
53. Payance A et al. Hepatocyte microvesicle levels improve prediction of mortality in patients with cirrhosis. Hepatology [published online ahead of print: March 30, 2018]; doi: 10.1002/hep.29903
54. Kubovcakova L et al. Differential effects of hydroxyurea and INC424 on mutant allele burden and myeloproliferative phenotype in a JAK2-V617F polycythemia vera mouse model. Blood 2013 ; 121 (7) : 1188- 1199.
55. Maschalidi S, Sepulveda FE, Garrigue A, Fischer A, de Saint Basile G. Therapeutic effect of JAK1/2 blockade on the manifestations of hemophagocytic lymphohistiocytosis in mice. Blood 2016;128(1):60-71.
56. Kou R, Shiroto T, Sartoretto JL, Michel T. Suppression of Gas synthesis by simvastatin treatment of vascular endothelial cells. J. Biol. Chem. 2012;287(4):2643-2651.
57. Lamrani L et al. Hemostatic disorders in a JAK2V617F-driven mouse model of myeloproliferative neoplasm. Blood 2014; 124(7): 1136-1145.
58. Poullet P, Carpentier S, Barillot E. myProMS, a web server for management and validation of mass spectrometry-based proteomic data. Proteomics 2007;7(15):2553-2556.
59. Valot B, Langella O, Nano E, Zivy M. MassChroQ: a versatile tool for mass spectrometry quantification. Proteomics 2011 ; 11(17):3572— 3577.
60. Vizcaino JA et al. 2016 update of the PRIDE database and its related tools. Nucleic Acids Res. 2016;44(Dl):D447-456.
Claims
1. A method of preventing the occurrence of cardiovascular event in a patient suffering from a myeloproliferative neoplasm comprising administering to the patient a therapeutically effective amount of a myeloperoxidase inhibitor.
2. The method of claim 1 wherein the patient suffers from polycythemia vera (PV), essential thrombocythemia (ET) or primary myelofibrosis (PMF).
3. The method of claim 1 wherein the patient harbors one mutation in JAK2.
4. The method of claim 3 wherein the mutation is the Jak2V617F mutation.
5. A method of determining whether a patient suffering from a myeloproliferative neoplasm is at risk of having a cardiovascular event comprising the steps of determining the level of microvesicles derived from Jak2V6I7F erythrocytes in a blood sample obtained from the patient wherein the level correlates with the risk of having a cardiovascular disease.
6. The method of claim 5 wherein the level of the microvesicles derived from Jak2V6I7F erythrocytes is compared to a predetermined reference value.
7. The method of claim 5 wherein the patient is treated with a statin when it is concluded that the patient has been determined as having a high risk of a cardiovascular event.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP19305975.5 | 2019-07-24 | ||
EP19305975 | 2019-07-24 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2021013942A1 true WO2021013942A1 (en) | 2021-01-28 |
Family
ID=67551313
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2020/070827 WO2021013942A1 (en) | 2019-07-24 | 2020-07-23 | Use of myeloperoxidase inhibitors for the treatment of cardiovascular diseases in patients suffering from myeloproliferative neoplasms |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2021013942A1 (en) |
Citations (13)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
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 |
US6107091A (en) | 1998-12-03 | 2000-08-22 | Isis Pharmaceuticals Inc. | Antisense inhibition of G-alpha-16 expression |
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 |
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 |
WO2003089430A1 (en) | 2002-04-19 | 2003-10-30 | Astrazeneca Ab | Thioxanthine derivatives as myeloperoxidase inhibitors |
WO2006062465A1 (en) | 2004-12-06 | 2006-06-15 | Astrazeneca Ab | Novel pyrrolo [3, 2-d] pyrimidin-4-one derivatives and their use in therapy |
US20120009608A1 (en) * | 2010-07-08 | 2012-01-12 | Azienda Ospedaliera Ospedali Riuniti Di Bergamo | Biomarker for the monitoring and prognosis of chronic myeloproliferative disorders |
US20120214825A1 (en) * | 2011-02-18 | 2012-08-23 | Vannucchi Alessandro M | mTOR/JAK INHIBITOR COMBINATION THERAPY |
WO2016178113A1 (en) * | 2015-05-05 | 2016-11-10 | Pfizer Inc. | 2-thiopyrimidinones |
US20190167680A1 (en) * | 2017-12-04 | 2019-06-06 | The Brigham And Women`S Hospital, Inc. | Inhibition of JAK-STAT Signaling Inhibits Formation of Neutrophil Extracellular Traps (NETs) |
-
2020
- 2020-07-23 WO PCT/EP2020/070827 patent/WO2021013942A1/en active Application Filing
Patent Citations (13)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
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 |
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 |
WO2003089430A1 (en) | 2002-04-19 | 2003-10-30 | Astrazeneca Ab | Thioxanthine derivatives as myeloperoxidase inhibitors |
WO2006062465A1 (en) | 2004-12-06 | 2006-06-15 | Astrazeneca Ab | Novel pyrrolo [3, 2-d] pyrimidin-4-one derivatives and their use in therapy |
US20120009608A1 (en) * | 2010-07-08 | 2012-01-12 | Azienda Ospedaliera Ospedali Riuniti Di Bergamo | Biomarker for the monitoring and prognosis of chronic myeloproliferative disorders |
US20120214825A1 (en) * | 2011-02-18 | 2012-08-23 | Vannucchi Alessandro M | mTOR/JAK INHIBITOR COMBINATION THERAPY |
WO2016178113A1 (en) * | 2015-05-05 | 2016-11-10 | Pfizer Inc. | 2-thiopyrimidinones |
US20190167680A1 (en) * | 2017-12-04 | 2019-06-06 | The Brigham And Women`S Hospital, Inc. | Inhibition of JAK-STAT Signaling Inhibits Formation of Neutrophil Extracellular Traps (NETs) |
Non-Patent Citations (65)
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Xia et al. | Autophagic adaptation to oxidative stress alters peritoneal residential macrophage survival and ovarian cancer metastasis | |
Gallo et al. | CCNE1 amplification is synthetic lethal with PKMYT1 kinase inhibition | |
Chen et al. | USP9X deubiquitinates ALDH1A3 and maintains mesenchymal identity in glioblastoma stem cells | |
Thomas et al. | The metabolic enzyme hexokinase 2 localizes to the nucleus in AML and normal haematopoietic stem and progenitor cells to maintain stemness | |
Kimishima et al. | Clonal hematopoiesis with JAK2V617F promotes pulmonary hypertension with ALK1 upregulation in lung neutrophils | |
Reddiconto et al. | Targeting of GSK3β promotes imatinib-mediated apoptosis in quiescent CD34+ chronic myeloid leukemia progenitors, preserving normal stem cells | |
US11208478B2 (en) | Methods of detecting and treating a tumor expressing pT346 PDK1 | |
US20210069218A1 (en) | Methods of controlling tumor bioenergetics networks | |
Liu et al. | R-loop accumulation in spliceosome mutant leukemias confers sensitivity to parp1 inhibition by triggering transcription–replication conflicts | |
Li et al. | Knockdown of LMNB1 inhibits the proliferation of lung adenocarcinoma cells by inducing DNA damage and cell senescence | |
CA3195753A1 (en) | Use of n-myristoyl transferase (nmt) inhibitors in the treatment of cancer, autoimmune disorders, and inflammatory disorders | |
US10828377B2 (en) | Method for determining presence or absence of suffering from malignant lymphoma or leukemia, and agent for treatment and/or prevention of leukemia | |
US20220062212A1 (en) | Compositions and Methods for Treating Patients Suffering from Glioma or Leukemia | |
Sharma et al. | Taurine from tumour niche drives glycolysis to promote leukaemogenesis | |
WO2021013942A1 (en) | Use of myeloperoxidase inhibitors for the treatment of cardiovascular diseases in patients suffering from myeloproliferative neoplasms | |
US20230158032A1 (en) | Pharmaceutical combination for the treatment of myeloproliferative neoplasms | |
WO2022255401A1 (en) | Disease marker expressed in association with abnormal erk-mapk pathway activation | |
Zhou et al. | USP14 modulates stem-like properties, tumorigenicity, and radiotherapy resistance in glioblastoma stem cells through stabilization of MST4-phosphorylated ALKBH5 | |
Orlikova-Boyer et al. | Antileukemic potential of methylated indolequinone MAC681 through immunogenic necroptosis and PARP1 degradation | |
Wei et al. | Plasma exosomes from patients with active thyroid-associated orbitopathy induce inflammation and fibrosis in orbital fibroblasts | |
US10213449B2 (en) | Compositions and methods for treating medulloblastoma | |
WO2021108927A1 (en) | Methods and compositions for treating cancers having f-box and wd-repeat protein 7 (fbxw7) alterations and/or cyclin l1 (ccnl1) gain or amplification | |
Toifl et al. | RAF1 kinase contributes to autophagic lysosome reformation | |
WO2019140257A1 (en) | Compositions and methods for characterizing and treating prostate cancer | |
Marinaccio | Myeloproliferative Neoplasms: From Biology to Targeted Therapeutics |
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: 20742291 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 20742291 Country of ref document: EP Kind code of ref document: A1 |