EP1686998A1 - Inhibition de la phosphodiesterase 9 comme traitement d'etats associes a l'obesite - Google Patents
Inhibition de la phosphodiesterase 9 comme traitement d'etats associes a l'obesiteInfo
- Publication number
- EP1686998A1 EP1686998A1 EP04769658A EP04769658A EP1686998A1 EP 1686998 A1 EP1686998 A1 EP 1686998A1 EP 04769658 A EP04769658 A EP 04769658A EP 04769658 A EP04769658 A EP 04769658A EP 1686998 A1 EP1686998 A1 EP 1686998A1
- Authority
- EP
- European Patent Office
- Prior art keywords
- pde9
- cell
- gene
- mouse
- cells
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Withdrawn
Links
- 101001117256 Drosophila melanogaster High affinity cGMP-specific 3',5'-cyclic phosphodiesterase 9A Proteins 0.000 title claims description 272
- 238000011282 treatment Methods 0.000 title abstract description 21
- 208000008589 Obesity Diseases 0.000 title description 10
- 235000020824 obesity Nutrition 0.000 title description 9
- 230000005764 inhibitory process Effects 0.000 title description 4
- 238000000034 method Methods 0.000 claims abstract description 70
- 241000699670 Mus sp. Species 0.000 claims abstract description 66
- 229940076380 PDE9 inhibitor Drugs 0.000 claims abstract description 48
- 241001465754 Metazoa Species 0.000 claims abstract description 40
- RVEJWGYZBXCGGM-DNVCBOLYSA-N chembl2179094 Chemical compound C([C@H]([C@@H](C1)C=2NC(=O)C=3C=NN(C=3N=2)C2CCOCC2)C)N1CC1=CC=CC=C1 RVEJWGYZBXCGGM-DNVCBOLYSA-N 0.000 claims abstract description 27
- 210000000577 adipose tissue Anatomy 0.000 claims abstract description 18
- 208000030814 Eating disease Diseases 0.000 claims abstract description 9
- 208000019454 Feeding and Eating disease Diseases 0.000 claims abstract description 9
- 235000014632 disordered eating Nutrition 0.000 claims abstract description 9
- 210000004027 cell Anatomy 0.000 claims description 173
- 241000699666 Mus <mouse, genus> Species 0.000 claims description 53
- 230000000694 effects Effects 0.000 claims description 41
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 27
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 25
- 229920001184 polypeptide Polymers 0.000 claims description 24
- 210000002459 blastocyst Anatomy 0.000 claims description 23
- 150000007523 nucleic acids Chemical class 0.000 claims description 16
- 235000009200 high fat diet Nutrition 0.000 claims description 14
- 241000124008 Mammalia Species 0.000 claims description 13
- 108091028043 Nucleic acid sequence Proteins 0.000 claims description 12
- 108020004707 nucleic acids Proteins 0.000 claims description 12
- 102000039446 nucleic acids Human genes 0.000 claims description 12
- 230000002829 reductive effect Effects 0.000 claims description 11
- 230000006798 recombination Effects 0.000 claims description 10
- 238000005215 recombination Methods 0.000 claims description 10
- 241001529936 Murinae Species 0.000 claims description 9
- 238000010363 gene targeting Methods 0.000 claims description 9
- 210000004962 mammalian cell Anatomy 0.000 claims description 9
- 230000001105 regulatory effect Effects 0.000 claims description 9
- 239000003112 inhibitor Substances 0.000 claims description 7
- 231100000272 reduced body weight Toxicity 0.000 claims description 6
- 229940124639 Selective inhibitor Drugs 0.000 claims description 4
- 101001072031 Drosophila melanogaster Dual 3',5'-cyclic-AMP and -GMP phosphodiesterase 11 Proteins 0.000 claims description 3
- 238000004519 manufacturing process Methods 0.000 claims description 3
- 108700008625 Reporter Genes Proteins 0.000 claims description 2
- 238000009395 breeding Methods 0.000 claims 1
- 230000001488 breeding effect Effects 0.000 claims 1
- 210000004102 animal cell Anatomy 0.000 abstract description 60
- 230000037396 body weight Effects 0.000 abstract description 23
- 206010033307 Overweight Diseases 0.000 abstract description 14
- 230000007423 decrease Effects 0.000 abstract description 11
- 235000013372 meat Nutrition 0.000 abstract description 6
- 235000013305 food Nutrition 0.000 abstract description 5
- 208000032841 Bulimia Diseases 0.000 abstract description 4
- 206010006550 Bulimia nervosa Diseases 0.000 abstract description 4
- 241000282887 Suidae Species 0.000 abstract description 4
- 208000014679 binge eating disease Diseases 0.000 abstract description 4
- 241000283690 Bos taurus Species 0.000 abstract description 3
- 241000287828 Gallus gallus Species 0.000 abstract description 3
- 235000013330 chicken meat Nutrition 0.000 abstract description 3
- 101100407341 Drosophila melanogaster Pde9 gene Proteins 0.000 abstract 2
- 239000013598 vector Substances 0.000 description 63
- 108090000623 proteins and genes Proteins 0.000 description 54
- 230000008685 targeting Effects 0.000 description 27
- 238000002744 homologous recombination Methods 0.000 description 25
- 230000006801 homologous recombination Effects 0.000 description 25
- 239000003550 marker Substances 0.000 description 25
- 150000001875 compounds Chemical class 0.000 description 22
- 230000014509 gene expression Effects 0.000 description 22
- 239000003795 chemical substances by application Substances 0.000 description 19
- 235000005911 diet Nutrition 0.000 description 18
- 230000037213 diet Effects 0.000 description 18
- 239000000203 mixture Substances 0.000 description 18
- 230000010354 integration Effects 0.000 description 16
- 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 15
- 238000012239 gene modification Methods 0.000 description 15
- 230000005017 genetic modification Effects 0.000 description 15
- 235000013617 genetically modified food Nutrition 0.000 description 15
- 239000008103 glucose Substances 0.000 description 15
- 102000004169 proteins and genes Human genes 0.000 description 15
- 210000004369 blood Anatomy 0.000 description 14
- 239000008280 blood Substances 0.000 description 14
- -1 anti-PDE9 antibodies Proteins 0.000 description 12
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 12
- 108091026890 Coding region Proteins 0.000 description 11
- 229940126062 Compound A Drugs 0.000 description 11
- NLDMNSXOCDLTTB-UHFFFAOYSA-N Heterophylliin A Natural products O1C2COC(=O)C3=CC(O)=C(O)C(O)=C3C3=C(O)C(O)=C(O)C=C3C(=O)OC2C(OC(=O)C=2C=C(O)C(O)=C(O)C=2)C(O)C1OC(=O)C1=CC(O)=C(O)C(O)=C1 NLDMNSXOCDLTTB-UHFFFAOYSA-N 0.000 description 11
- 230000003247 decreasing effect Effects 0.000 description 11
- 239000002552 dosage form Substances 0.000 description 10
- 210000001519 tissue Anatomy 0.000 description 10
- 108020004414 DNA Proteins 0.000 description 9
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 9
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 9
- 238000003556 assay Methods 0.000 description 9
- 239000003814 drug Substances 0.000 description 9
- 238000009472 formulation Methods 0.000 description 9
- 230000036541 health Effects 0.000 description 9
- 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 8
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 8
- 229940079593 drug Drugs 0.000 description 8
- 230000006870 function Effects 0.000 description 8
- 238000009396 hybridization Methods 0.000 description 8
- 230000001939 inductive effect Effects 0.000 description 8
- 230000004048 modification Effects 0.000 description 8
- 238000012986 modification Methods 0.000 description 8
- 239000000546 pharmaceutical excipient Substances 0.000 description 8
- 150000003839 salts Chemical class 0.000 description 8
- 108700028369 Alleles Proteins 0.000 description 7
- 108090001050 Phosphoric Diester Hydrolases Proteins 0.000 description 7
- 102000004861 Phosphoric Diester Hydrolases Human genes 0.000 description 7
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 7
- 239000002299 complementary DNA Substances 0.000 description 7
- 235000012631 food intake Nutrition 0.000 description 7
- 210000001161 mammalian embryo Anatomy 0.000 description 7
- 239000002773 nucleotide Substances 0.000 description 7
- 125000003729 nucleotide group Chemical group 0.000 description 7
- 238000011160 research Methods 0.000 description 7
- 229920000858 Cyclodextrin Polymers 0.000 description 6
- 102000004190 Enzymes Human genes 0.000 description 6
- 108090000790 Enzymes Proteins 0.000 description 6
- 102000004877 Insulin Human genes 0.000 description 6
- 108090001061 Insulin Proteins 0.000 description 6
- 108700019146 Transgenes Proteins 0.000 description 6
- 239000002775 capsule Substances 0.000 description 6
- 201000010099 disease Diseases 0.000 description 6
- 210000002257 embryonic structure Anatomy 0.000 description 6
- 210000004602 germ cell Anatomy 0.000 description 6
- 230000001965 increasing effect Effects 0.000 description 6
- 238000003780 insertion Methods 0.000 description 6
- 230000037431 insertion Effects 0.000 description 6
- 229940125396 insulin Drugs 0.000 description 6
- 108020004999 messenger RNA Proteins 0.000 description 6
- 230000035772 mutation Effects 0.000 description 6
- 238000003752 polymerase chain reaction Methods 0.000 description 6
- 239000000651 prodrug Substances 0.000 description 6
- 229940002612 prodrug Drugs 0.000 description 6
- 210000001082 somatic cell Anatomy 0.000 description 6
- 238000010561 standard procedure Methods 0.000 description 6
- 210000000130 stem cell Anatomy 0.000 description 6
- 239000000126 substance Substances 0.000 description 6
- 239000000758 substrate Substances 0.000 description 6
- 230000002123 temporal effect Effects 0.000 description 6
- UFTFJSFQGQCHQW-UHFFFAOYSA-N triformin Chemical compound O=COCC(OC=O)COC=O UFTFJSFQGQCHQW-UHFFFAOYSA-N 0.000 description 6
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 6
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 5
- 241000282412 Homo Species 0.000 description 5
- 206010020772 Hypertension Diseases 0.000 description 5
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 5
- 108010091086 Recombinases Proteins 0.000 description 5
- 102000018120 Recombinases Human genes 0.000 description 5
- 239000000556 agonist Substances 0.000 description 5
- QQKNSPHAFATFNQ-UHFFFAOYSA-N darglitazone Chemical compound CC=1OC(C=2C=CC=CC=2)=NC=1CCC(=O)C(C=C1)=CC=C1CC1SC(=O)NC1=O QQKNSPHAFATFNQ-UHFFFAOYSA-N 0.000 description 5
- 229950006689 darglitazone Drugs 0.000 description 5
- 238000012217 deletion Methods 0.000 description 5
- 230000037430 deletion Effects 0.000 description 5
- 238000010353 genetic engineering Methods 0.000 description 5
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 description 5
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 description 5
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 5
- UFVKGYZPFZQRLF-UHFFFAOYSA-N hydroxypropyl methyl cellulose Chemical compound OC1C(O)C(OC)OC(CO)C1OC1C(O)C(O)C(OC2C(C(O)C(OC3C(C(O)C(O)C(CO)O3)O)C(CO)O2)O)C(CO)O1 UFVKGYZPFZQRLF-UHFFFAOYSA-N 0.000 description 5
- 230000004060 metabolic process Effects 0.000 description 5
- 239000013612 plasmid Substances 0.000 description 5
- 229920001223 polyethylene glycol Polymers 0.000 description 5
- 230000008569 process Effects 0.000 description 5
- 239000000523 sample Substances 0.000 description 5
- 239000000243 solution Substances 0.000 description 5
- 235000019786 weight gain Nutrition 0.000 description 5
- 108010051219 Cre recombinase Proteins 0.000 description 4
- 241000283984 Rodentia Species 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- 238000002105 Southern blotting Methods 0.000 description 4
- 229920002472 Starch Polymers 0.000 description 4
- 239000000443 aerosol Substances 0.000 description 4
- 238000004458 analytical method Methods 0.000 description 4
- 239000000883 anti-obesity agent Substances 0.000 description 4
- 229940125710 antiobesity agent Drugs 0.000 description 4
- 238000004113 cell culture Methods 0.000 description 4
- 239000003085 diluting agent Substances 0.000 description 4
- 210000001671 embryonic stem cell Anatomy 0.000 description 4
- 230000037406 food intake Effects 0.000 description 4
- IXZISFNWUWKBOM-ARQDHWQXSA-N fructosamine Chemical compound NC[C@@]1(O)OC[C@@H](O)[C@@H](O)[C@@H]1O IXZISFNWUWKBOM-ARQDHWQXSA-N 0.000 description 4
- 239000000499 gel Substances 0.000 description 4
- 210000002216 heart Anatomy 0.000 description 4
- 238000003018 immunoassay Methods 0.000 description 4
- 210000004072 lung Anatomy 0.000 description 4
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 4
- 230000001404 mediated effect Effects 0.000 description 4
- 238000002493 microarray Methods 0.000 description 4
- 238000002703 mutagenesis Methods 0.000 description 4
- 231100000350 mutagenesis Toxicity 0.000 description 4
- AHLBNYSZXLDEJQ-FWEHEUNISA-N orlistat Chemical compound CCCCCCCCCCC[C@H](OC(=O)[C@H](CC(C)C)NC=O)C[C@@H]1OC(=O)[C@H]1CCCCCC AHLBNYSZXLDEJQ-FWEHEUNISA-N 0.000 description 4
- 229960001243 orlistat Drugs 0.000 description 4
- 235000019271 petrolatum Nutrition 0.000 description 4
- 210000001778 pluripotent stem cell Anatomy 0.000 description 4
- 239000000843 powder Substances 0.000 description 4
- 238000002360 preparation method Methods 0.000 description 4
- 230000009467 reduction Effects 0.000 description 4
- 239000011780 sodium chloride Substances 0.000 description 4
- 241000894007 species Species 0.000 description 4
- 229940032147 starch Drugs 0.000 description 4
- 239000008107 starch Substances 0.000 description 4
- 235000019698 starch Nutrition 0.000 description 4
- 208000001608 teratocarcinoma Diseases 0.000 description 4
- 208000001072 type 2 diabetes mellitus Diseases 0.000 description 4
- WVDDGKGOMKODPV-UHFFFAOYSA-N Benzyl alcohol Chemical compound OCC1=CC=CC=C1 WVDDGKGOMKODPV-UHFFFAOYSA-N 0.000 description 3
- 238000002965 ELISA Methods 0.000 description 3
- 241000588724 Escherichia coli Species 0.000 description 3
- 108091060211 Expressed sequence tag Proteins 0.000 description 3
- 108010010803 Gelatin Proteins 0.000 description 3
- 102000016267 Leptin Human genes 0.000 description 3
- 108010092277 Leptin Proteins 0.000 description 3
- 241001494479 Pecora Species 0.000 description 3
- 239000002202 Polyethylene glycol Substances 0.000 description 3
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 3
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 3
- 239000008186 active pharmaceutical agent Substances 0.000 description 3
- 230000004075 alteration Effects 0.000 description 3
- 239000005557 antagonist Substances 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- 230000008901 benefit Effects 0.000 description 3
- 230000015572 biosynthetic process Effects 0.000 description 3
- 229960002802 bromocriptine Drugs 0.000 description 3
- OZVBMTJYIDMWIL-AYFBDAFISA-N bromocriptine Chemical compound C1=CC(C=2[C@H](N(C)C[C@@H](C=2)C(=O)N[C@]2(C(=O)N3[C@H](C(N4CCC[C@H]4[C@]3(O)O2)=O)CC(C)C)C(C)C)C2)=C3C2=C(Br)NC3=C1 OZVBMTJYIDMWIL-AYFBDAFISA-N 0.000 description 3
- 235000012000 cholesterol Nutrition 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 238000002591 computed tomography Methods 0.000 description 3
- 230000000875 corresponding effect Effects 0.000 description 3
- 125000004122 cyclic group Chemical group 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 229940088679 drug related substance Drugs 0.000 description 3
- 238000004520 electroporation Methods 0.000 description 3
- 238000005516 engineering process Methods 0.000 description 3
- 230000007613 environmental effect Effects 0.000 description 3
- 230000002255 enzymatic effect Effects 0.000 description 3
- 239000012634 fragment Substances 0.000 description 3
- 239000008273 gelatin Substances 0.000 description 3
- 229920000159 gelatin Polymers 0.000 description 3
- 235000019322 gelatine Nutrition 0.000 description 3
- 235000011852 gelatine desserts Nutrition 0.000 description 3
- 230000002710 gonadal effect Effects 0.000 description 3
- 210000005260 human cell Anatomy 0.000 description 3
- 229920000639 hydroxypropylmethylcellulose acetate succinate Polymers 0.000 description 3
- 239000004615 ingredient Substances 0.000 description 3
- 239000008101 lactose Substances 0.000 description 3
- 229960001375 lactose Drugs 0.000 description 3
- NRYBAZVQPHGZNS-ZSOCWYAHSA-N leptin Chemical compound O=C([C@H](CO)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)CNC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](N)CC(C)C)CCSC)N1CCC[C@H]1C(=O)NCC(=O)N[C@@H](CS)C(O)=O NRYBAZVQPHGZNS-ZSOCWYAHSA-N 0.000 description 3
- 230000013011 mating Effects 0.000 description 3
- 239000002674 ointment Substances 0.000 description 3
- 239000008177 pharmaceutical agent Substances 0.000 description 3
- 239000008194 pharmaceutical composition Substances 0.000 description 3
- 230000000541 pulsatile effect Effects 0.000 description 3
- 229940044551 receptor antagonist Drugs 0.000 description 3
- 239000002464 receptor antagonist Substances 0.000 description 3
- 238000003757 reverse transcription PCR Methods 0.000 description 3
- UNAANXDKBXWMLN-UHFFFAOYSA-N sibutramine Chemical compound C=1C=C(Cl)C=CC=1C1(C(N(C)C)CC(C)C)CCC1 UNAANXDKBXWMLN-UHFFFAOYSA-N 0.000 description 3
- 229960004425 sibutramine Drugs 0.000 description 3
- 239000012453 solvate Substances 0.000 description 3
- 239000002904 solvent Substances 0.000 description 3
- 239000007921 spray Substances 0.000 description 3
- 238000006467 substitution reaction Methods 0.000 description 3
- 239000000725 suspension Substances 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 238000012546 transfer Methods 0.000 description 3
- 230000009261 transgenic effect Effects 0.000 description 3
- 150000003626 triacylglycerols Chemical class 0.000 description 3
- 210000004291 uterus Anatomy 0.000 description 3
- 230000035899 viability Effects 0.000 description 3
- 239000013603 viral vector Substances 0.000 description 3
- KWGRBVOPPLSCSI-WPRPVWTQSA-N (-)-ephedrine Chemical compound CN[C@@H](C)[C@H](O)C1=CC=CC=C1 KWGRBVOPPLSCSI-WPRPVWTQSA-N 0.000 description 2
- ZISBIXCLKMNALM-UHFFFAOYSA-N 9-(3-methylbutan-2-yl)-2-[[2-(2-morpholin-4-ylethoxy)phenyl]methyl]-3h-purin-6-one Chemical compound CC(C)C(C)N1C=NC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCCN1CCOCC1 ZISBIXCLKMNALM-UHFFFAOYSA-N 0.000 description 2
- LRFVTYWOQMYALW-UHFFFAOYSA-N 9H-xanthine Chemical compound O=C1NC(=O)NC2=C1NC=N2 LRFVTYWOQMYALW-UHFFFAOYSA-N 0.000 description 2
- 208000004611 Abdominal Obesity Diseases 0.000 description 2
- 102000007469 Actins Human genes 0.000 description 2
- 108010085238 Actins Proteins 0.000 description 2
- 108020005544 Antisense RNA Proteins 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 241000271566 Aves Species 0.000 description 2
- 238000011740 C57BL/6 mouse Methods 0.000 description 2
- VTYYLEPIZMXCLO-UHFFFAOYSA-L Calcium carbonate Chemical compound [Ca+2].[O-]C([O-])=O VTYYLEPIZMXCLO-UHFFFAOYSA-L 0.000 description 2
- 241000282472 Canis lupus familiaris Species 0.000 description 2
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 2
- 208000024172 Cardiovascular disease Diseases 0.000 description 2
- 108090000994 Catalytic RNA Proteins 0.000 description 2
- 102000053642 Catalytic RNA Human genes 0.000 description 2
- 206010065941 Central obesity Diseases 0.000 description 2
- 101710150887 Cholecystokinin A Proteins 0.000 description 2
- 108020004705 Codon Proteins 0.000 description 2
- 108020004635 Complementary DNA Proteins 0.000 description 2
- 229920002785 Croscarmellose sodium Polymers 0.000 description 2
- 241000701022 Cytomegalovirus Species 0.000 description 2
- 206010059866 Drug resistance Diseases 0.000 description 2
- 239000001856 Ethyl cellulose Substances 0.000 description 2
- ZZSNKZQZMQGXPY-UHFFFAOYSA-N Ethyl cellulose Chemical compound CCOCC1OC(OC)C(OCC)C(OCC)C1OC1C(O)C(O)C(OC)C(CO)O1 ZZSNKZQZMQGXPY-UHFFFAOYSA-N 0.000 description 2
- 241000282326 Felis catus Species 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 229920002527 Glycogen Polymers 0.000 description 2
- 241000288105 Grus Species 0.000 description 2
- NTYJJOPFIAHURM-UHFFFAOYSA-N Histamine Chemical compound NCCC1=CN=CN1 NTYJJOPFIAHURM-UHFFFAOYSA-N 0.000 description 2
- 108010033040 Histones Proteins 0.000 description 2
- 101150003028 Hprt1 gene Proteins 0.000 description 2
- 229920002153 Hydroxypropyl cellulose Polymers 0.000 description 2
- 208000031773 Insulin resistance syndrome Diseases 0.000 description 2
- ZQISRDCJNBUVMM-UHFFFAOYSA-N L-Histidinol Natural products OCC(N)CC1=CN=CN1 ZQISRDCJNBUVMM-UHFFFAOYSA-N 0.000 description 2
- ZQISRDCJNBUVMM-YFKPBYRVSA-N L-histidinol Chemical compound OC[C@@H](N)CC1=CNC=N1 ZQISRDCJNBUVMM-YFKPBYRVSA-N 0.000 description 2
- 208000001145 Metabolic Syndrome Diseases 0.000 description 2
- 102100031545 Microsomal triglyceride transfer protein large subunit Human genes 0.000 description 2
- 206010028980 Neoplasm Diseases 0.000 description 2
- 108091034117 Oligonucleotide Proteins 0.000 description 2
- 239000004264 Petrolatum Substances 0.000 description 2
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 239000004147 Sorbitan trioleate Substances 0.000 description 2
- PRXRUNOAOLTIEF-ADSICKODSA-N Sorbitan trioleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OC[C@@H](OC(=O)CCCCCCC\C=C/CCCCCCCC)[C@H]1OC[C@H](O)[C@H]1OC(=O)CCCCCCC\C=C/CCCCCCCC PRXRUNOAOLTIEF-ADSICKODSA-N 0.000 description 2
- 239000004098 Tetracycline Substances 0.000 description 2
- 208000035199 Tetraploidy Diseases 0.000 description 2
- 102000006601 Thymidine Kinase Human genes 0.000 description 2
- 108020004440 Thymidine kinase Proteins 0.000 description 2
- 201000000690 abdominal obesity-metabolic syndrome Diseases 0.000 description 2
- ZUAAPNNKRHMPKG-UHFFFAOYSA-N acetic acid;butanedioic acid;methanol;propane-1,2-diol Chemical compound OC.CC(O)=O.CC(O)CO.OC(=O)CCC(O)=O ZUAAPNNKRHMPKG-UHFFFAOYSA-N 0.000 description 2
- OIRDTQYFTABQOQ-KQYNXXCUSA-N adenosine Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O OIRDTQYFTABQOQ-KQYNXXCUSA-N 0.000 description 2
- 150000001413 amino acids Chemical class 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 239000007864 aqueous solution Substances 0.000 description 2
- 238000003491 array Methods 0.000 description 2
- 230000036772 blood pressure Effects 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- 210000001612 chondrocyte Anatomy 0.000 description 2
- 238000010367 cloning Methods 0.000 description 2
- 239000003184 complementary RNA Substances 0.000 description 2
- 238000010276 construction Methods 0.000 description 2
- 238000013270 controlled release Methods 0.000 description 2
- 239000006071 cream Substances 0.000 description 2
- 229960001681 croscarmellose sodium Drugs 0.000 description 2
- 235000010947 crosslinked sodium carboxy methyl cellulose Nutrition 0.000 description 2
- KWGRBVOPPLSCSI-UHFFFAOYSA-N d-ephedrine Natural products CNC(C)C(O)C1=CC=CC=C1 KWGRBVOPPLSCSI-UHFFFAOYSA-N 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 230000007812 deficiency Effects 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 206010012601 diabetes mellitus Diseases 0.000 description 2
- 230000004069 differentiation Effects 0.000 description 2
- 208000035475 disorder Diseases 0.000 description 2
- 238000001962 electrophoresis Methods 0.000 description 2
- 239000003995 emulsifying agent Substances 0.000 description 2
- 210000002889 endothelial cell Anatomy 0.000 description 2
- 239000003623 enhancer Substances 0.000 description 2
- 235000019325 ethyl cellulose Nutrition 0.000 description 2
- 229920001249 ethyl cellulose Polymers 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 210000002950 fibroblast Anatomy 0.000 description 2
- 239000000796 flavoring agent Substances 0.000 description 2
- 230000009368 gene silencing by RNA Effects 0.000 description 2
- 230000002068 genetic effect Effects 0.000 description 2
- 210000001654 germ layer Anatomy 0.000 description 2
- 235000011187 glycerol Nutrition 0.000 description 2
- 229940096919 glycogen Drugs 0.000 description 2
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 2
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 2
- 150000004677 hydrates Chemical class 0.000 description 2
- 235000010977 hydroxypropyl cellulose Nutrition 0.000 description 2
- 239000001863 hydroxypropyl cellulose Substances 0.000 description 2
- FDGQSTZJBFJUBT-UHFFFAOYSA-N hypoxanthine Chemical compound O=C1NC=NC2=C1NC=N2 FDGQSTZJBFJUBT-UHFFFAOYSA-N 0.000 description 2
- 230000002452 interceptive effect Effects 0.000 description 2
- 229940039781 leptin Drugs 0.000 description 2
- 231100000518 lethal Toxicity 0.000 description 2
- 230000001665 lethal effect Effects 0.000 description 2
- 150000002632 lipids Chemical class 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 239000006210 lotion Substances 0.000 description 2
- 239000000314 lubricant Substances 0.000 description 2
- 235000019359 magnesium stearate Nutrition 0.000 description 2
- 239000011159 matrix material Substances 0.000 description 2
- 230000002503 metabolic effect Effects 0.000 description 2
- 239000002207 metabolite Substances 0.000 description 2
- 238000000520 microinjection Methods 0.000 description 2
- 239000002480 mineral oil Substances 0.000 description 2
- 235000010446 mineral oil Nutrition 0.000 description 2
- 239000003607 modifier Substances 0.000 description 2
- 210000003205 muscle Anatomy 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 210000002569 neuron Anatomy 0.000 description 2
- 229910052757 nitrogen Inorganic materials 0.000 description 2
- 210000000287 oocyte Anatomy 0.000 description 2
- 210000000056 organ Anatomy 0.000 description 2
- 238000007911 parenteral administration Methods 0.000 description 2
- 230000001776 parthenogenetic effect Effects 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 229940066842 petrolatum Drugs 0.000 description 2
- 230000000144 pharmacologic effect Effects 0.000 description 2
- 238000001556 precipitation Methods 0.000 description 2
- 239000003380 propellant Substances 0.000 description 2
- 238000011002 quantification Methods 0.000 description 2
- 238000003127 radioimmunoassay Methods 0.000 description 2
- 229940044601 receptor agonist Drugs 0.000 description 2
- 239000000018 receptor agonist Substances 0.000 description 2
- 238000010839 reverse transcription Methods 0.000 description 2
- 108091092562 ribozyme Proteins 0.000 description 2
- HFHDHCJBZVLPGP-UHFFFAOYSA-N schardinger α-dextrin Chemical compound O1C(C(C2O)O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC(C(O)C2O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC2C(O)C(O)C1OC2CO HFHDHCJBZVLPGP-UHFFFAOYSA-N 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- 210000002027 skeletal muscle Anatomy 0.000 description 2
- 210000003491 skin Anatomy 0.000 description 2
- 150000003384 small molecules Chemical class 0.000 description 2
- 235000019337 sorbitan trioleate Nutrition 0.000 description 2
- 229960000391 sorbitan trioleate Drugs 0.000 description 2
- 230000010473 stable expression Effects 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000000375 suspending agent Substances 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 210000001550 testis Anatomy 0.000 description 2
- 229960002180 tetracycline Drugs 0.000 description 2
- 229930101283 tetracycline Natural products 0.000 description 2
- 235000019364 tetracycline Nutrition 0.000 description 2
- 150000003522 tetracyclines Chemical class 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- WYWHKKSPHMUBEB-UHFFFAOYSA-N tioguanine Chemical compound N1C(N)=NC(=S)C2=C1N=CN2 WYWHKKSPHMUBEB-UHFFFAOYSA-N 0.000 description 2
- 238000012250 transgenic expression Methods 0.000 description 2
- 238000013519 translation Methods 0.000 description 2
- 231100000588 tumorigenic Toxicity 0.000 description 2
- 230000000381 tumorigenic effect Effects 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- 230000003442 weekly effect Effects 0.000 description 2
- DIGQNXIGRZPYDK-WKSCXVIASA-N (2R)-6-amino-2-[[2-[[(2S)-2-[[2-[[(2R)-2-[[(2S)-2-[[(2R,3S)-2-[[2-[[(2S)-2-[[2-[[(2S)-2-[[(2S)-2-[[(2R)-2-[[(2S,3S)-2-[[(2R)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[2-[[(2S)-2-[[(2R)-2-[[2-[[2-[[2-[(2-amino-1-hydroxyethylidene)amino]-3-carboxy-1-hydroxypropylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxybutylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1,5-dihydroxy-5-iminopentylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxybutylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxyethylidene]amino]hexanoic acid Chemical compound C[C@@H]([C@@H](C(=N[C@@H](CS)C(=N[C@@H](C)C(=N[C@@H](CO)C(=NCC(=N[C@@H](CCC(=N)O)C(=NC(CS)C(=N[C@H]([C@H](C)O)C(=N[C@H](CS)C(=N[C@H](CO)C(=NCC(=N[C@H](CS)C(=NCC(=N[C@H](CCCCN)C(=O)O)O)O)O)O)O)O)O)O)O)O)O)O)O)N=C([C@H](CS)N=C([C@H](CO)N=C([C@H](CO)N=C([C@H](C)N=C(CN=C([C@H](CO)N=C([C@H](CS)N=C(CN=C(C(CS)N=C(C(CC(=O)O)N=C(CN)O)O)O)O)O)O)O)O)O)O)O)O DIGQNXIGRZPYDK-WKSCXVIASA-N 0.000 description 1
- LNAZSHAWQACDHT-XIYTZBAFSA-N (2r,3r,4s,5r,6s)-4,5-dimethoxy-2-(methoxymethyl)-3-[(2s,3r,4s,5r,6r)-3,4,5-trimethoxy-6-(methoxymethyl)oxan-2-yl]oxy-6-[(2r,3r,4s,5r,6r)-4,5,6-trimethoxy-2-(methoxymethyl)oxan-3-yl]oxyoxane Chemical compound CO[C@@H]1[C@@H](OC)[C@H](OC)[C@@H](COC)O[C@H]1O[C@H]1[C@H](OC)[C@@H](OC)[C@H](O[C@H]2[C@@H]([C@@H](OC)[C@H](OC)O[C@@H]2COC)OC)O[C@@H]1COC LNAZSHAWQACDHT-XIYTZBAFSA-N 0.000 description 1
- WRIDQFICGBMAFQ-UHFFFAOYSA-N (E)-8-Octadecenoic acid Natural products CCCCCCCCCC=CCCCCCCC(O)=O WRIDQFICGBMAFQ-UHFFFAOYSA-N 0.000 description 1
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- YFMFNYKEUDLDTL-UHFFFAOYSA-N 1,1,1,2,3,3,3-heptafluoropropane Chemical compound FC(F)(F)C(F)C(F)(F)F YFMFNYKEUDLDTL-UHFFFAOYSA-N 0.000 description 1
- LVGUZGTVOIAKKC-UHFFFAOYSA-N 1,1,1,2-tetrafluoroethane Chemical compound FCC(F)(F)F LVGUZGTVOIAKKC-UHFFFAOYSA-N 0.000 description 1
- DDMOUSALMHHKOS-UHFFFAOYSA-N 1,2-dichloro-1,1,2,2-tetrafluoroethane Chemical compound FC(F)(Cl)C(F)(F)Cl DDMOUSALMHHKOS-UHFFFAOYSA-N 0.000 description 1
- BLJIGEMJBJIDIM-UHFFFAOYSA-N 1-[2-[2-[(7-oxo-3-propan-2-yl-2,4-dihydropyrazolo[4,3-d]pyrimidin-5-yl)methyl]phenoxy]acetyl]pyrrolidine-2-carboxylic acid Chemical compound CC(C)C1=NNC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCC(=O)N1CCCC1C(O)=O BLJIGEMJBJIDIM-UHFFFAOYSA-N 0.000 description 1
- OKMWKBLSFKFYGZ-UHFFFAOYSA-N 1-behenoylglycerol Chemical compound CCCCCCCCCCCCCCCCCCCCCC(=O)OCC(O)CO OKMWKBLSFKFYGZ-UHFFFAOYSA-N 0.000 description 1
- DKTPMOWCDYVXMF-UHFFFAOYSA-N 2-[[2-(2-morpholin-4-ylethoxy)phenyl]methyl]-9-(oxolan-2-ylmethyl)-3h-purin-6-one Chemical compound N=1C=2N(CC3OCCC3)C=NC=2C(=O)NC=1CC1=CC=CC=C1OCCN1CCOCC1 DKTPMOWCDYVXMF-UHFFFAOYSA-N 0.000 description 1
- FMYHIADPRHYCNE-UHFFFAOYSA-N 2-[[2-(2-morpholin-4-ylethoxy)phenyl]methyl]-9-(oxolan-3-yl)-3h-purin-6-one Chemical compound N=1C=2N(C3COCC3)C=NC=2C(=O)NC=1CC1=CC=CC=C1OCCN1CCOCC1 FMYHIADPRHYCNE-UHFFFAOYSA-N 0.000 description 1
- UFLFUHCXWBQZLS-UHFFFAOYSA-N 2-[[2-(2-morpholin-4-ylethoxy)phenyl]methyl]-9-pentan-3-yl-3h-purin-6-one Chemical compound CCC(CC)N1C=NC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCCN1CCOCC1 UFLFUHCXWBQZLS-UHFFFAOYSA-N 0.000 description 1
- RSUVEZGNQWCKFS-UHFFFAOYSA-N 2-[[2-(2-morpholin-4-ylethoxy)phenyl]methyl]-9-propan-2-yl-3h-purin-6-one Chemical compound CC(C)N1C=NC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCCN1CCOCC1 RSUVEZGNQWCKFS-UHFFFAOYSA-N 0.000 description 1
- LEACJMVNYZDSKR-UHFFFAOYSA-N 2-octyldodecan-1-ol Chemical compound CCCCCCCCCCC(CO)CCCCCCCC LEACJMVNYZDSKR-UHFFFAOYSA-N 0.000 description 1
- LQJBNNIYVWPHFW-UHFFFAOYSA-N 20:1omega9c fatty acid Natural products CCCCCCCCCCC=CCCCCCCCC(O)=O LQJBNNIYVWPHFW-UHFFFAOYSA-N 0.000 description 1
- LFLCFQVVSCAUHI-UHFFFAOYSA-N 3-cyclopentyl-5-[[2-(2-morpholin-4-ylethoxy)phenyl]methyl]-2,4-dihydropyrazolo[4,3-d]pyrimidin-7-one Chemical compound N=1C=2C(C3CCCC3)=NNC=2C(=O)NC=1CC1=CC=CC=C1OCCN1CCOCC1 LFLCFQVVSCAUHI-UHFFFAOYSA-N 0.000 description 1
- FZOCMUYBRUIGHI-UHFFFAOYSA-N 3-cyclopentyl-5-[[5-fluoro-2-(2-morpholin-4-ylethoxy)phenyl]methyl]-2,4-dihydropyrazolo[4,3-d]pyrimidin-7-one Chemical compound N=1C=2C(C3CCCC3)=NNC=2C(=O)NC=1CC1=CC(F)=CC=C1OCCN1CCOCC1 FZOCMUYBRUIGHI-UHFFFAOYSA-N 0.000 description 1
- BCXQFEMQSMIDLW-UHFFFAOYSA-N 3-propan-2-yl-5-[[2-(2-pyrrolidin-1-ylethoxy)phenyl]methyl]-2,4-dihydropyrazolo[4,3-d]pyrimidin-7-one Chemical compound CC(C)C1=NNC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCCN1CCCC1 BCXQFEMQSMIDLW-UHFFFAOYSA-N 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
- HETHZASKFZMWLF-UHFFFAOYSA-N 5-[[2-(2-morpholin-4-yl-2-oxoethoxy)phenyl]methyl]-3-propan-2-yl-2,4-dihydropyrazolo[4,3-d]pyrimidin-7-one Chemical compound CC(C)C1=NNC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCC(=O)N1CCOCC1 HETHZASKFZMWLF-UHFFFAOYSA-N 0.000 description 1
- DHNHXAUERSTPRA-UHFFFAOYSA-N 5-[[2-(2-morpholin-4-ylethoxy)cyclohexyl]methyl]-3-propan-2-yl-2,4-dihydropyrazolo[4,3-d]pyrimidin-7-one Chemical compound CC(C)C1=NNC(C(N2)=O)=C1N=C2CC1CCCCC1OCCN1CCOCC1 DHNHXAUERSTPRA-UHFFFAOYSA-N 0.000 description 1
- DQQIKKSUASWOMX-UHFFFAOYSA-N 5-[[2-(2-morpholin-4-ylethoxy)phenyl]methyl]-3-propan-2-yl-2,4-dihydropyrazolo[4,3-d]pyrimidin-7-one Chemical compound CC(C)C1=NNC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCCN1CCOCC1 DQQIKKSUASWOMX-UHFFFAOYSA-N 0.000 description 1
- WFZJBMDOKXMQNS-UHFFFAOYSA-N 5-[[2-(2-morpholin-4-ylethoxy)phenyl]methyl]-3-pyridin-3-yl-1,4-dihydropyrazolo[4,3-d]pyrimidin-7-one Chemical compound N=1C=2C(C=3C=NC=CC=3)=NNC=2C(=O)NC=1CC1=CC=CC=C1OCCN1CCOCC1 WFZJBMDOKXMQNS-UHFFFAOYSA-N 0.000 description 1
- DCPSLQUYKGGVIP-UHFFFAOYSA-N 5-[[2-(2-oxo-2-piperazin-1-ylethoxy)phenyl]methyl]-3-propan-2-yl-2,4-dihydropyrazolo[4,3-d]pyrimidin-7-one;2,2,2-trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F.CC(C)C=1NN=C(C(N=2)=O)C=1NC=2CC1=CC=CC=C1OCC(=O)N1CCNCC1 DCPSLQUYKGGVIP-UHFFFAOYSA-N 0.000 description 1
- SRAMCTHPVUSCIP-UHFFFAOYSA-N 5-[[2-(2-oxo-2-pyrrolidin-1-ylethoxy)phenyl]methyl]-3-propan-2-yl-2,4-dihydropyrazolo[4,3-d]pyrimidin-7-one Chemical compound CC(C)C1=NNC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCC(=O)N1CCCC1 SRAMCTHPVUSCIP-UHFFFAOYSA-N 0.000 description 1
- WNMAQZGAMUXFGF-UHFFFAOYSA-N 5-[[2-[2-(3-chloromorpholin-4-yl)ethoxy]phenyl]methyl]-3-propan-2-yl-2,4-dihydropyrazolo[4,3-d]pyrimidin-7-one Chemical compound CC(C)C1=NNC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCCN1CCOCC1Cl WNMAQZGAMUXFGF-UHFFFAOYSA-N 0.000 description 1
- OEGMYACNENBDAK-UHFFFAOYSA-N 5-[[2-[2-(diethylamino)ethoxy]phenyl]methyl]-3-propan-2-yl-2,4-dihydropyrazolo[4,3-d]pyrimidin-7-one Chemical compound CCN(CC)CCOC1=CC=CC=C1CC(NC1=O)=NC2=C1NN=C2C(C)C OEGMYACNENBDAK-UHFFFAOYSA-N 0.000 description 1
- RJOXFJDOUQJOMQ-UHFFFAOYSA-N 6-sulfanylidene-3,7-dihydropurin-2-one Chemical compound S=C1NC(=O)NC2=C1NC=N2 RJOXFJDOUQJOMQ-UHFFFAOYSA-N 0.000 description 1
- BOXCRKCQNZDZIQ-UHFFFAOYSA-N 9-(2,4-dimethylpentan-3-yl)-2-[[2-(2-morpholin-4-ylethoxy)phenyl]methyl]-3h-purin-6-one Chemical compound CC(C)C(C(C)C)N1C=NC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCCN1CCOCC1 BOXCRKCQNZDZIQ-UHFFFAOYSA-N 0.000 description 1
- YVOLLHBANQWEEW-UHFFFAOYSA-N 9-(2-methylbutyl)-2-[[2-(2-morpholin-4-ylethoxy)phenyl]methyl]-3h-purin-6-one Chemical compound CCC(C)CN1C=NC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCCN1CCOCC1 YVOLLHBANQWEEW-UHFFFAOYSA-N 0.000 description 1
- QSBYPNXLFMSGKH-UHFFFAOYSA-N 9-Heptadecensaeure Natural products CCCCCCCC=CCCCCCCCC(O)=O QSBYPNXLFMSGKH-UHFFFAOYSA-N 0.000 description 1
- KLXLCGOALWQKCO-UHFFFAOYSA-N 9-cyclopentyl-2-[[2-(2-morpholin-4-ylethoxy)phenyl]methyl]-3h-purin-6-one Chemical compound N=1C=2N(C3CCCC3)C=NC=2C(=O)NC=1CC1=CC=CC=C1OCCN1CCOCC1 KLXLCGOALWQKCO-UHFFFAOYSA-N 0.000 description 1
- WBZFUFAFFUEMEI-UHFFFAOYSA-M Acesulfame k Chemical compound [K+].CC1=CC(=O)[N-]S(=O)(=O)O1 WBZFUFAFFUEMEI-UHFFFAOYSA-M 0.000 description 1
- NIXOWILDQLNWCW-UHFFFAOYSA-N Acrylic acid Chemical compound OC(=O)C=C NIXOWILDQLNWCW-UHFFFAOYSA-N 0.000 description 1
- 102000054930 Agouti-Related Human genes 0.000 description 1
- 101710127426 Agouti-related protein Proteins 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- PQSUYGKTWSAVDQ-ZVIOFETBSA-N Aldosterone Chemical compound C([C@@]1([C@@H](C(=O)CO)CC[C@H]1[C@@H]1CC2)C=O)[C@H](O)[C@@H]1[C@]1(C)C2=CC(=O)CC1 PQSUYGKTWSAVDQ-ZVIOFETBSA-N 0.000 description 1
- PQSUYGKTWSAVDQ-UHFFFAOYSA-N Aldosterone Natural products C1CC2C3CCC(C(=O)CO)C3(C=O)CC(O)C2C2(C)C1=CC(=O)CC2 PQSUYGKTWSAVDQ-UHFFFAOYSA-N 0.000 description 1
- 229920001450 Alpha-Cyclodextrin Polymers 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- 206010002091 Anaesthesia Diseases 0.000 description 1
- 101710095342 Apolipoprotein B Proteins 0.000 description 1
- 102100040202 Apolipoprotein B-100 Human genes 0.000 description 1
- 102000018616 Apolipoproteins B Human genes 0.000 description 1
- 108010027006 Apolipoproteins B Proteins 0.000 description 1
- 102100021569 Apoptosis regulator Bcl-2 Human genes 0.000 description 1
- 108010011485 Aspartame Proteins 0.000 description 1
- 108010006654 Bleomycin Proteins 0.000 description 1
- 208000031648 Body Weight Changes Diseases 0.000 description 1
- 108010051479 Bombesin Proteins 0.000 description 1
- 102000013585 Bombesin Human genes 0.000 description 1
- 239000002126 C01EB10 - Adenosine Substances 0.000 description 1
- 102100024316 Calcium/calmodulin-dependent 3',5'-cyclic nucleotide phosphodiesterase 1A Human genes 0.000 description 1
- 102100024317 Calcium/calmodulin-dependent 3',5'-cyclic nucleotide phosphodiesterase 1C Human genes 0.000 description 1
- 229940122820 Cannabinoid receptor antagonist Drugs 0.000 description 1
- 241000701489 Cauliflower mosaic virus Species 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 229920000623 Cellulose acetate phthalate Polymers 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 1
- 208000017667 Chronic Disease Diseases 0.000 description 1
- 108020004638 Circular DNA Proteins 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- 108010080611 Cytosine Deaminase Proteins 0.000 description 1
- 102000000311 Cytosine Deaminase Human genes 0.000 description 1
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 1
- 208000019505 Deglutition disease Diseases 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- 239000004338 Dichlorodifluoromethane Substances 0.000 description 1
- 101100189582 Dictyostelium discoideum pdeD gene Proteins 0.000 description 1
- 101100351286 Dictyostelium discoideum pdeE gene Proteins 0.000 description 1
- 101100135859 Dictyostelium discoideum regA gene Proteins 0.000 description 1
- 108010053187 Diphtheria Toxin Proteins 0.000 description 1
- 102000016607 Diphtheria Toxin Human genes 0.000 description 1
- 101001117089 Drosophila melanogaster Calcium/calmodulin-dependent 3',5'-cyclic nucleotide phosphodiesterase 1 Proteins 0.000 description 1
- 208000032928 Dyslipidaemia Diseases 0.000 description 1
- YQYJSBFKSSDGFO-UHFFFAOYSA-N Epihygromycin Natural products OC1C(O)C(C(=O)C)OC1OC(C(=C1)O)=CC=C1C=C(C)C(=O)NC1C(O)C(O)C2OCOC2C1O YQYJSBFKSSDGFO-UHFFFAOYSA-N 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 241001452028 Escherichia coli DH1 Species 0.000 description 1
- 241001646716 Escherichia coli K-12 Species 0.000 description 1
- JIGUQPWFLRLWPJ-UHFFFAOYSA-N Ethyl acrylate Chemical compound CCOC(=O)C=C JIGUQPWFLRLWPJ-UHFFFAOYSA-N 0.000 description 1
- 108700024394 Exon Proteins 0.000 description 1
- 108090000331 Firefly luciferases Proteins 0.000 description 1
- 102400001370 Galanin Human genes 0.000 description 1
- 101800002068 Galanin Proteins 0.000 description 1
- 108010016122 Ghrelin Receptors Proteins 0.000 description 1
- 208000002705 Glucose Intolerance Diseases 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 1
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 1
- 102100039256 Growth hormone secretagogue receptor type 1 Human genes 0.000 description 1
- 102100021519 Hemoglobin subunit beta Human genes 0.000 description 1
- 108091005904 Hemoglobin subunit beta Proteins 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- 208000009889 Herpes Simplex Diseases 0.000 description 1
- 102100024233 High affinity cAMP-specific 3',5'-cyclic phosphodiesterase 7A Human genes 0.000 description 1
- 102100024228 High affinity cAMP-specific and IBMX-insensitive 3',5'-cyclic phosphodiesterase 8A Human genes 0.000 description 1
- 102100024229 High affinity cAMP-specific and IBMX-insensitive 3',5'-cyclic phosphodiesterase 8B Human genes 0.000 description 1
- 101710145025 High affinity cAMP-specific and IBMX-insensitive 3',5'-cyclic phosphodiesterase 8B Proteins 0.000 description 1
- 239000004705 High-molecular-weight polyethylene Substances 0.000 description 1
- 101000928179 Homo sapiens Agouti-related protein Proteins 0.000 description 1
- 101000971171 Homo sapiens Apoptosis regulator Bcl-2 Proteins 0.000 description 1
- 101001117044 Homo sapiens Calcium/calmodulin-dependent 3',5'-cyclic nucleotide phosphodiesterase 1A Proteins 0.000 description 1
- 101001117094 Homo sapiens Calcium/calmodulin-dependent 3',5'-cyclic nucleotide phosphodiesterase 1C Proteins 0.000 description 1
- 101001117267 Homo sapiens High affinity cAMP-specific 3',5'-cyclic phosphodiesterase 7A Proteins 0.000 description 1
- 101001117261 Homo sapiens High affinity cAMP-specific and IBMX-insensitive 3',5'-cyclic phosphodiesterase 8A Proteins 0.000 description 1
- 101001098805 Homo sapiens cAMP-specific 3',5'-cyclic phosphodiesterase 4A Proteins 0.000 description 1
- 101000988424 Homo sapiens cAMP-specific 3',5'-cyclic phosphodiesterase 4B Proteins 0.000 description 1
- 101000988423 Homo sapiens cAMP-specific 3',5'-cyclic phosphodiesterase 4C Proteins 0.000 description 1
- 101000988419 Homo sapiens cAMP-specific 3',5'-cyclic phosphodiesterase 4D Proteins 0.000 description 1
- 101001117266 Homo sapiens cAMP-specific 3',5'-cyclic phosphodiesterase 7B Proteins 0.000 description 1
- 101001098818 Homo sapiens cGMP-inhibited 3',5'-cyclic phosphodiesterase A Proteins 0.000 description 1
- 101001098812 Homo sapiens cGMP-inhibited 3',5'-cyclic phosphodiesterase B Proteins 0.000 description 1
- 101100321817 Human parvovirus B19 (strain HV) 7.5K gene Proteins 0.000 description 1
- 208000031226 Hyperlipidaemia Diseases 0.000 description 1
- UGQMRVRMYYASKQ-UHFFFAOYSA-N Hypoxanthine nucleoside Natural products OC1C(O)C(CO)OC1N1C(NC=NC2=O)=C2N=C1 UGQMRVRMYYASKQ-UHFFFAOYSA-N 0.000 description 1
- 206010056997 Impaired fasting glucose Diseases 0.000 description 1
- 208000015580 Increased body weight Diseases 0.000 description 1
- 206010022489 Insulin Resistance Diseases 0.000 description 1
- 240000007472 Leucaena leucocephala Species 0.000 description 1
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 1
- 229940127470 Lipase Inhibitors Drugs 0.000 description 1
- 208000017170 Lipid metabolism disease Diseases 0.000 description 1
- WHXSMMKQMYFTQS-UHFFFAOYSA-N Lithium Chemical compound [Li] WHXSMMKQMYFTQS-UHFFFAOYSA-N 0.000 description 1
- 108010000410 MSH receptor Proteins 0.000 description 1
- 240000003183 Manihot esculenta Species 0.000 description 1
- 235000016735 Manihot esculenta subsp esculenta Nutrition 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 102400001132 Melanin-concentrating hormone Human genes 0.000 description 1
- 101800002739 Melanin-concentrating hormone Proteins 0.000 description 1
- 102100034216 Melanocyte-stimulating hormone receptor Human genes 0.000 description 1
- 101710151321 Melanostatin Proteins 0.000 description 1
- 235000006679 Mentha X verticillata Nutrition 0.000 description 1
- 235000002899 Mentha suaveolens Nutrition 0.000 description 1
- 235000001636 Mentha x rotundifolia Nutrition 0.000 description 1
- 102000003792 Metallothionein Human genes 0.000 description 1
- 108090000157 Metallothionein Proteins 0.000 description 1
- VVQNEPGJFQJSBK-UHFFFAOYSA-N Methyl methacrylate Chemical compound COC(=O)C(C)=C VVQNEPGJFQJSBK-UHFFFAOYSA-N 0.000 description 1
- 206010027525 Microalbuminuria Diseases 0.000 description 1
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 1
- 241000282339 Mustela Species 0.000 description 1
- 101000909851 Mycobacterium tuberculosis (strain ATCC 25618 / H37Rv) cAMP/cGMP dual specificity phosphodiesterase Rv0805 Proteins 0.000 description 1
- WHNWPMSKXPGLAX-UHFFFAOYSA-N N-Vinyl-2-pyrrolidone Chemical compound C=CN1CCCC1=O WHNWPMSKXPGLAX-UHFFFAOYSA-N 0.000 description 1
- AHLBNYSZXLDEJQ-UHFFFAOYSA-N N-formyl-L-leucylester Natural products CCCCCCCCCCCC(OC(=O)C(CC(C)C)NC=O)CC1OC(=O)C1CCCCCC AHLBNYSZXLDEJQ-UHFFFAOYSA-N 0.000 description 1
- 229930193140 Neomycin Natural products 0.000 description 1
- 108010025020 Nerve Growth Factor Proteins 0.000 description 1
- 102000007072 Nerve Growth Factors Human genes 0.000 description 1
- 102000030937 Neuromedin U receptor Human genes 0.000 description 1
- 108010002741 Neuromedin U receptor Proteins 0.000 description 1
- 102400000064 Neuropeptide Y Human genes 0.000 description 1
- 108091092724 Noncoding DNA Proteins 0.000 description 1
- 238000000636 Northern blotting Methods 0.000 description 1
- 239000005642 Oleic acid Substances 0.000 description 1
- ZQPPMHVWECSIRJ-UHFFFAOYSA-N Oleic acid Natural products CCCCCCCCC=CCCCCCCCC(O)=O ZQPPMHVWECSIRJ-UHFFFAOYSA-N 0.000 description 1
- 108700020796 Oncogene Proteins 0.000 description 1
- 108010058765 Oncogene Protein pp60(v-src) Proteins 0.000 description 1
- 108700026244 Open Reading Frames Proteins 0.000 description 1
- 229940123730 Orexin receptor antagonist Drugs 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 238000012408 PCR amplification Methods 0.000 description 1
- 238000010222 PCR analysis Methods 0.000 description 1
- 101150098694 PDE5A gene Proteins 0.000 description 1
- 102000012288 Phosphopyruvate Hydratase Human genes 0.000 description 1
- 108010022181 Phosphopyruvate Hydratase Proteins 0.000 description 1
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 1
- 101100082606 Plasmodium falciparum (isolate 3D7) PDEbeta gene Proteins 0.000 description 1
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 1
- 229920001214 Polysorbate 60 Polymers 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 206010038997 Retroviral infections Diseases 0.000 description 1
- 108010039491 Ricin Proteins 0.000 description 1
- 101100135860 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) PDE2 gene Proteins 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- 229920002125 Sokalan® Polymers 0.000 description 1
- 244000061456 Solanum tuberosum Species 0.000 description 1
- 235000002595 Solanum tuberosum Nutrition 0.000 description 1
- HVUMOYIDDBPOLL-XWVZOOPGSA-N Sorbitan monostearate Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@@H](O)[C@H]1OC[C@H](O)[C@H]1O HVUMOYIDDBPOLL-XWVZOOPGSA-N 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- 241000862969 Stella Species 0.000 description 1
- 208000006011 Stroke Diseases 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 241000282898 Sus scrofa Species 0.000 description 1
- 239000000150 Sympathomimetic Substances 0.000 description 1
- 230000006044 T cell activation Effects 0.000 description 1
- 108010017842 Telomerase Proteins 0.000 description 1
- PHYFQTYBJUILEZ-UHFFFAOYSA-N Trioleoylglycerol Natural products CCCCCCCCC=CCCCCCCCC(=O)OCC(OC(=O)CCCCCCCC=CCCCCCCCC)COC(=O)CCCCCCCC=CCCCCCCCC PHYFQTYBJUILEZ-UHFFFAOYSA-N 0.000 description 1
- 241000700618 Vaccinia virus Species 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- TVXBFESIOXBWNM-UHFFFAOYSA-N Xylitol Natural products OCCC(O)C(O)C(O)CCO TVXBFESIOXBWNM-UHFFFAOYSA-N 0.000 description 1
- 240000008042 Zea mays Species 0.000 description 1
- 235000005824 Zea mays ssp. parviglumis Nutrition 0.000 description 1
- 235000002017 Zea mays subsp mays Nutrition 0.000 description 1
- VSYMNDBTCKIDLT-UHFFFAOYSA-N [2-(carbamoyloxymethyl)-2-ethylbutyl] carbamate Chemical compound NC(=O)OCC(CC)(CC)COC(N)=O VSYMNDBTCKIDLT-UHFFFAOYSA-N 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 235000010358 acesulfame potassium Nutrition 0.000 description 1
- 229960004998 acesulfame potassium Drugs 0.000 description 1
- 239000000619 acesulfame-K Substances 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 230000006978 adaptation Effects 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 229960005305 adenosine Drugs 0.000 description 1
- 210000001789 adipocyte Anatomy 0.000 description 1
- 239000000048 adrenergic agonist Substances 0.000 description 1
- 229940126157 adrenergic receptor agonist Drugs 0.000 description 1
- 230000004931 aggregating effect Effects 0.000 description 1
- 230000002776 aggregation Effects 0.000 description 1
- 238000004220 aggregation Methods 0.000 description 1
- 230000008484 agonism Effects 0.000 description 1
- 229960002478 aldosterone Drugs 0.000 description 1
- 230000037005 anaesthesia Effects 0.000 description 1
- 229940125709 anorectic agent Drugs 0.000 description 1
- 230000000692 anti-sense effect Effects 0.000 description 1
- 239000000074 antisense oligonucleotide Substances 0.000 description 1
- 238000012230 antisense oligonucleotides Methods 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 239000002830 appetite depressant Substances 0.000 description 1
- 239000007900 aqueous suspension Substances 0.000 description 1
- 206010003246 arthritis Diseases 0.000 description 1
- 239000000605 aspartame Substances 0.000 description 1
- IAOZJIPTCAWIRG-QWRGUYRKSA-N aspartame Chemical compound OC(=O)C[C@H](N)C(=O)N[C@H](C(=O)OC)CC1=CC=CC=C1 IAOZJIPTCAWIRG-QWRGUYRKSA-N 0.000 description 1
- 235000010357 aspartame Nutrition 0.000 description 1
- 229960003438 aspartame Drugs 0.000 description 1
- 235000019445 benzyl alcohol Nutrition 0.000 description 1
- 102000005936 beta-Galactosidase Human genes 0.000 description 1
- 108010005774 beta-Galactosidase Proteins 0.000 description 1
- 229910002056 binary alloy Inorganic materials 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 229920001222 biopolymer Polymers 0.000 description 1
- 229960001561 bleomycin Drugs 0.000 description 1
- OYVAGSVQBOHSSS-UAPAGMARSA-O bleomycin A2 Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCC[S+](C)C)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C OYVAGSVQBOHSSS-UAPAGMARSA-O 0.000 description 1
- 230000004579 body weight change Effects 0.000 description 1
- DNDCVAGJPBKION-DOPDSADYSA-N bombesin Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(N)=O)NC(=O)CNC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](CC=1NC2=CC=CC=C2C=1)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)CNC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H]1NC(=O)CC1)C(C)C)C1=CN=CN1 DNDCVAGJPBKION-DOPDSADYSA-N 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 102100037092 cAMP-specific 3',5'-cyclic phosphodiesterase 4A Human genes 0.000 description 1
- 102100029168 cAMP-specific 3',5'-cyclic phosphodiesterase 4B Human genes 0.000 description 1
- 102100029169 cAMP-specific 3',5'-cyclic phosphodiesterase 4C Human genes 0.000 description 1
- 102100029170 cAMP-specific 3',5'-cyclic phosphodiesterase 4D Human genes 0.000 description 1
- 102100024232 cAMP-specific 3',5'-cyclic phosphodiesterase 7B Human genes 0.000 description 1
- 102100037093 cGMP-inhibited 3',5'-cyclic phosphodiesterase A Human genes 0.000 description 1
- 102100037094 cGMP-inhibited 3',5'-cyclic phosphodiesterase B Human genes 0.000 description 1
- 102100029175 cGMP-specific 3',5'-cyclic phosphodiesterase Human genes 0.000 description 1
- 229910000019 calcium carbonate Inorganic materials 0.000 description 1
- 229960003563 calcium carbonate Drugs 0.000 description 1
- XAAHAAMILDNBPS-UHFFFAOYSA-L calcium hydrogenphosphate dihydrate Chemical compound O.O.[Ca+2].OP([O-])([O-])=O XAAHAAMILDNBPS-UHFFFAOYSA-L 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229960001714 calcium phosphate Drugs 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 244000309466 calf Species 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 239000003536 cannabinoid receptor antagonist Substances 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 229960001631 carbomer Drugs 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 229910002092 carbon dioxide Inorganic materials 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 210000004413 cardiac myocyte Anatomy 0.000 description 1
- 239000004203 carnauba wax Substances 0.000 description 1
- 235000013869 carnauba wax Nutrition 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 239000004359 castor oil Substances 0.000 description 1
- 235000019438 castor oil Nutrition 0.000 description 1
- 230000003197 catalytic effect Effects 0.000 description 1
- 230000022131 cell cycle Effects 0.000 description 1
- 230000003833 cell viability Effects 0.000 description 1
- 235000010980 cellulose Nutrition 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 229920002301 cellulose acetate Polymers 0.000 description 1
- 229940081734 cellulose acetate phthalate Drugs 0.000 description 1
- 229940081733 cetearyl alcohol Drugs 0.000 description 1
- 238000012412 chemical coupling Methods 0.000 description 1
- 230000002759 chromosomal effect Effects 0.000 description 1
- 230000001886 ciliary effect Effects 0.000 description 1
- 229960004106 citric acid Drugs 0.000 description 1
- 235000015165 citric acid Nutrition 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 210000001072 colon Anatomy 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 238000004040 coloring Methods 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 238000012875 competitive assay Methods 0.000 description 1
- 230000002860 competitive effect Effects 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 238000010668 complexation reaction Methods 0.000 description 1
- 238000012790 confirmation Methods 0.000 description 1
- 235000020940 control diet Nutrition 0.000 description 1
- 229920001577 copolymer Polymers 0.000 description 1
- 235000005822 corn Nutrition 0.000 description 1
- 208000029078 coronary artery disease Diseases 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 229940109239 creatinine Drugs 0.000 description 1
- DDRJAANPRJIHGJ-UHFFFAOYSA-N creatinine Natural products CN1CC(=O)NC1=N DDRJAANPRJIHGJ-UHFFFAOYSA-N 0.000 description 1
- 229960000913 crospovidone Drugs 0.000 description 1
- 238000004132 cross linking Methods 0.000 description 1
- 229940097362 cyclodextrins Drugs 0.000 description 1
- 238000004163 cytometry Methods 0.000 description 1
- 230000006735 deficit Effects 0.000 description 1
- 239000007933 dermal patch Substances 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 235000019700 dicalcium phosphate Nutrition 0.000 description 1
- 229940095079 dicalcium phosphate anhydrous Drugs 0.000 description 1
- PXBRQCKWGAHEHS-UHFFFAOYSA-N dichlorodifluoromethane Chemical compound FC(F)(Cl)Cl PXBRQCKWGAHEHS-UHFFFAOYSA-N 0.000 description 1
- 235000019404 dichlorodifluoromethane Nutrition 0.000 description 1
- 229940042935 dichlorodifluoromethane Drugs 0.000 description 1
- 229940087091 dichlorotetrafluoroethane Drugs 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- 239000006185 dispersion Substances 0.000 description 1
- 238000004090 dissolution Methods 0.000 description 1
- 229940052760 dopamine agonists Drugs 0.000 description 1
- 239000003136 dopamine receptor stimulating agent Substances 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 238000009510 drug design Methods 0.000 description 1
- 238000007876 drug discovery Methods 0.000 description 1
- 229940112141 dry powder inhaler Drugs 0.000 description 1
- 238000001035 drying Methods 0.000 description 1
- 238000010410 dusting Methods 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 210000003981 ectoderm Anatomy 0.000 description 1
- 230000002526 effect on cardiovascular system Effects 0.000 description 1
- 230000001804 emulsifying effect Effects 0.000 description 1
- 239000008387 emulsifying waxe Substances 0.000 description 1
- 210000003890 endocrine cell Anatomy 0.000 description 1
- 210000001900 endoderm Anatomy 0.000 description 1
- 230000002357 endometrial effect Effects 0.000 description 1
- 210000003979 eosinophil Anatomy 0.000 description 1
- 229960002179 ephedrine Drugs 0.000 description 1
- 201000010063 epididymitis Diseases 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 210000003013 erythroid precursor cell Anatomy 0.000 description 1
- 150000002170 ethers Chemical class 0.000 description 1
- MVPICKVDHDWCJQ-UHFFFAOYSA-N ethyl 3-pyrrolidin-1-ylpropanoate Chemical compound CCOC(=O)CCN1CCCC1 MVPICKVDHDWCJQ-UHFFFAOYSA-N 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 210000002907 exocrine cell Anatomy 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- XRECTZIEBJDKEO-UHFFFAOYSA-N flucytosine Chemical compound NC1=NC(=O)NC=C1F XRECTZIEBJDKEO-UHFFFAOYSA-N 0.000 description 1
- 229960004413 flucytosine Drugs 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 235000003599 food sweetener Nutrition 0.000 description 1
- 231100000221 frame shift mutation induction Toxicity 0.000 description 1
- 230000037433 frameshift Effects 0.000 description 1
- 229910021485 fumed silica Inorganic materials 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 208000020694 gallbladder disease Diseases 0.000 description 1
- IRSCQMHQWWYFCW-UHFFFAOYSA-N ganciclovir Chemical compound O=C1NC(N)=NC2=C1N=CN2COC(CO)CO IRSCQMHQWWYFCW-UHFFFAOYSA-N 0.000 description 1
- 229960002963 ganciclovir Drugs 0.000 description 1
- 239000007789 gas Substances 0.000 description 1
- 238000012817 gel-diffusion technique Methods 0.000 description 1
- 239000007903 gelatin capsule Substances 0.000 description 1
- 210000004907 gland Anatomy 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 239000003877 glucagon like peptide 1 receptor agonist Substances 0.000 description 1
- 229940124750 glucocorticoid receptor agonist Drugs 0.000 description 1
- 230000004190 glucose uptake Effects 0.000 description 1
- ZEMPKEQAKRGZGQ-XOQCFJPHSA-N glycerol triricinoleate Natural products CCCCCC[C@@H](O)CC=CCCCCCCCC(=O)OC[C@@H](COC(=O)CCCCCCCC=CC[C@@H](O)CCCCCC)OC(=O)CCCCCCCC=CC[C@H](O)CCCCCC ZEMPKEQAKRGZGQ-XOQCFJPHSA-N 0.000 description 1
- 229940049654 glyceryl behenate Drugs 0.000 description 1
- 229960002449 glycine Drugs 0.000 description 1
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 1
- 238000005469 granulation Methods 0.000 description 1
- 230000003179 granulation Effects 0.000 description 1
- 230000005484 gravity Effects 0.000 description 1
- 239000005090 green fluorescent protein Substances 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 229960002897 heparin Drugs 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 229960001340 histamine Drugs 0.000 description 1
- 210000003630 histaminocyte Anatomy 0.000 description 1
- 239000003667 hormone antagonist Substances 0.000 description 1
- 102000055839 human AGRP Human genes 0.000 description 1
- 150000005828 hydrofluoroalkanes Chemical class 0.000 description 1
- 239000000017 hydrogel Substances 0.000 description 1
- 230000007062 hydrolysis Effects 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- 229920003132 hydroxypropyl methylcellulose phthalate Polymers 0.000 description 1
- 229940031704 hydroxypropyl methylcellulose phthalate Drugs 0.000 description 1
- 208000006575 hypertriglyceridemia Diseases 0.000 description 1
- 238000010191 image analysis Methods 0.000 description 1
- 239000012729 immediate-release (IR) formulation Substances 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000000951 immunodiffusion Effects 0.000 description 1
- 238000000760 immunoelectrophoresis Methods 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 238000007901 in situ hybridization Methods 0.000 description 1
- 238000003017 in situ immunoassay Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 230000003914 insulin secretion Effects 0.000 description 1
- 230000004155 insulin signaling pathway Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000011835 investigation Methods 0.000 description 1
- QXJSBBXBKPUZAA-UHFFFAOYSA-N isooleic acid Natural products CCCCCCCC=CCCCCCCCCC(O)=O QXJSBBXBKPUZAA-UHFFFAOYSA-N 0.000 description 1
- 210000002510 keratinocyte Anatomy 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 238000011813 knockout mouse model Methods 0.000 description 1
- 102000005861 leptin receptors Human genes 0.000 description 1
- 108010019813 leptin receptors Proteins 0.000 description 1
- 231100000225 lethality Toxicity 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 229940057995 liquid paraffin Drugs 0.000 description 1
- 229910052744 lithium Inorganic materials 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 238000007726 management method Methods 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 210000003593 megakaryocyte Anatomy 0.000 description 1
- ORRDHOMWDPJSNL-UHFFFAOYSA-N melanin concentrating hormone Chemical compound N1C(=O)C(C(C)C)NC(=O)C(CCCNC(N)=N)NC(=O)CNC(=O)C(C(C)C)NC(=O)C(CCSC)NC(=O)C(NC(=O)C(CCCNC(N)=N)NC(=O)C(NC(=O)C(NC(=O)C(N)CC(O)=O)C(C)O)CCSC)CSSCC(C(=O)NC(CC=2C3=CC=CC=C3NC=2)C(=O)NC(CCC(O)=O)C(=O)NC(C(C)C)C(O)=O)NC(=O)C2CCCN2C(=O)C(CCCNC(N)=N)NC(=O)C1CC1=CC=C(O)C=C1 ORRDHOMWDPJSNL-UHFFFAOYSA-N 0.000 description 1
- HEBKCHPVOIAQTA-UHFFFAOYSA-N meso ribitol Natural products OCC(O)C(O)C(O)CO HEBKCHPVOIAQTA-UHFFFAOYSA-N 0.000 description 1
- 210000003716 mesoderm Anatomy 0.000 description 1
- 210000005033 mesothelial cell Anatomy 0.000 description 1
- 229920003145 methacrylic acid copolymer Polymers 0.000 description 1
- 229940117841 methacrylic acid copolymer Drugs 0.000 description 1
- MYWUZJCMWCOHBA-VIFPVBQESA-N methamphetamine Chemical compound CN[C@@H](C)CC1=CC=CC=C1 MYWUZJCMWCOHBA-VIFPVBQESA-N 0.000 description 1
- ZDYPMGBHMKRSFD-UHFFFAOYSA-N methyl 1-[2-[2-[(7-oxo-3-propan-2-yl-2,4-dihydropyrazolo[4,3-d]pyrimidin-5-yl)methyl]phenoxy]acetyl]pyrrolidine-2-carboxylate Chemical compound COC(=O)C1CCCN1C(=O)COC1=CC=CC=C1CC(NC1=O)=NC2=C1NN=C2C(C)C ZDYPMGBHMKRSFD-UHFFFAOYSA-N 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 235000010981 methylcellulose Nutrition 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 238000010603 microCT Methods 0.000 description 1
- 239000008108 microcrystalline cellulose Substances 0.000 description 1
- 229940016286 microcrystalline cellulose Drugs 0.000 description 1
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 1
- 108010038232 microsomal triglyceride transfer protein Proteins 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- SLZIZIJTGAYEKK-CIJSCKBQSA-N molport-023-220-247 Chemical compound C([C@@H](C(=O)N[C@@H](C)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=1N=CNC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1N=CNC=1)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C)C(N)=O)NC(=O)[C@H]1N(CCC1)C(=O)CNC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)CN)[C@@H](C)O)C1=CNC=N1 SLZIZIJTGAYEKK-CIJSCKBQSA-N 0.000 description 1
- 230000000407 monoamine reuptake Effects 0.000 description 1
- 210000000472 morula Anatomy 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 210000000663 muscle cell Anatomy 0.000 description 1
- 210000004165 myocardium Anatomy 0.000 description 1
- CUPRRXXFEMOLLE-UHFFFAOYSA-N n-[2-(dimethylamino)ethyl]-2-[2-[(7-oxo-3-propan-2-yl-2,4-dihydropyrazolo[4,3-d]pyrimidin-5-yl)methyl]phenoxy]acetamide Chemical compound CC(C)C1=NNC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCC(=O)NCCN(C)C CUPRRXXFEMOLLE-UHFFFAOYSA-N 0.000 description 1
- SEYCIBHCEGUQNB-UHFFFAOYSA-N n-[2-[(7-oxo-3-propan-2-yl-2,4-dihydropyrazolo[4,3-d]pyrimidin-5-yl)methyl]cyclohexyl]-2-pyrrolidin-1-ylacetamide Chemical compound CC(C)C1=NNC(C(N2)=O)=C1N=C2CC1CCCCC1NC(=O)CN1CCCC1 SEYCIBHCEGUQNB-UHFFFAOYSA-N 0.000 description 1
- 229960004927 neomycin Drugs 0.000 description 1
- 230000009826 neoplastic cell growth Effects 0.000 description 1
- 230000001537 neural effect Effects 0.000 description 1
- 239000003900 neurotrophic factor Substances 0.000 description 1
- 230000036963 noncompetitive effect Effects 0.000 description 1
- 238000007899 nucleic acid hybridization Methods 0.000 description 1
- URPYMXQQVHTUDU-OFGSCBOVSA-N nucleopeptide y Chemical compound C([C@@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(N)=O)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CCCCN)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@@H](N)CC=1C=CC(O)=CC=1)C1=CC=C(O)C=C1 URPYMXQQVHTUDU-OFGSCBOVSA-N 0.000 description 1
- GLDOVTGHNKAZLK-UHFFFAOYSA-N octadecan-1-ol Chemical compound CCCCCCCCCCCCCCCCCCO GLDOVTGHNKAZLK-UHFFFAOYSA-N 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 1
- ZQPPMHVWECSIRJ-KTKRTIGZSA-N oleic acid Chemical compound CCCCCCCC\C=C/CCCCCCCC(O)=O ZQPPMHVWECSIRJ-KTKRTIGZSA-N 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 230000008520 organization Effects 0.000 description 1
- 201000008482 osteoarthritis Diseases 0.000 description 1
- 210000004409 osteocyte Anatomy 0.000 description 1
- 230000001151 other effect Effects 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 235000019809 paraffin wax Nutrition 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 101150037969 pde-6 gene Proteins 0.000 description 1
- PNJWIWWMYCMZRO-UHFFFAOYSA-N pent‐4‐en‐2‐one Natural products CC(=O)CC=C PNJWIWWMYCMZRO-UHFFFAOYSA-N 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 239000002831 pharmacologic agent Substances 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 235000010989 polyoxyethylene sorbitan monostearate Nutrition 0.000 description 1
- 239000001818 polyoxyethylene sorbitan monostearate Substances 0.000 description 1
- 229920002503 polyoxyethylene-polyoxypropylene Polymers 0.000 description 1
- 229940113124 polysorbate 60 Drugs 0.000 description 1
- 229920000523 polyvinylpolypyrrolidone Polymers 0.000 description 1
- 235000013809 polyvinylpolypyrrolidone Nutrition 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 238000011176 pooling Methods 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 201000009104 prediabetes syndrome Diseases 0.000 description 1
- 230000002028 premature Effects 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000002335 preservative effect Effects 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 230000001902 propagating effect Effects 0.000 description 1
- 235000013772 propylene glycol Nutrition 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 238000013197 protein A assay Methods 0.000 description 1
- 229960003908 pseudoephedrine Drugs 0.000 description 1
- KWGRBVOPPLSCSI-WCBMZHEXSA-N pseudoephedrine Chemical compound CN[C@@H](C)[C@@H](O)C1=CC=CC=C1 KWGRBVOPPLSCSI-WCBMZHEXSA-N 0.000 description 1
- 230000005180 public health Effects 0.000 description 1
- 230000002685 pulmonary effect Effects 0.000 description 1
- WAJNANMQOPCIPO-UHFFFAOYSA-N pyrazolo[4,3-d]pyrimidin-7-one Chemical compound O=C1N=CN=C2C=NN=C12 WAJNANMQOPCIPO-UHFFFAOYSA-N 0.000 description 1
- 239000002469 receptor inverse agonist Substances 0.000 description 1
- 238000004064 recycling Methods 0.000 description 1
- 230000004648 relaxation of smooth muscle Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- JZCPYUJPEARBJL-UHFFFAOYSA-N rimonabant Chemical compound CC=1C(C(=O)NN2CCCCC2)=NN(C=2C(=CC(Cl)=CC=2)Cl)C=1C1=CC=C(Cl)C=C1 JZCPYUJPEARBJL-UHFFFAOYSA-N 0.000 description 1
- 229960003015 rimonabant Drugs 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 230000008786 sensory perception of smell Effects 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 150000004760 silicates Chemical class 0.000 description 1
- 239000000377 silicon dioxide Substances 0.000 description 1
- 235000012239 silicon dioxide Nutrition 0.000 description 1
- 229960001866 silicon dioxide Drugs 0.000 description 1
- 210000002363 skeletal muscle cell Anatomy 0.000 description 1
- 201000002859 sleep apnea Diseases 0.000 description 1
- 210000000813 small intestine Anatomy 0.000 description 1
- 210000002460 smooth muscle Anatomy 0.000 description 1
- 210000000329 smooth muscle myocyte Anatomy 0.000 description 1
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- 239000001509 sodium citrate Substances 0.000 description 1
- 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 1
- 229960001790 sodium citrate Drugs 0.000 description 1
- 229940080313 sodium starch Drugs 0.000 description 1
- 239000008109 sodium starch glycolate Substances 0.000 description 1
- 229940079832 sodium starch glycolate Drugs 0.000 description 1
- 229920003109 sodium starch glycolate Polymers 0.000 description 1
- 229940045902 sodium stearyl fumarate Drugs 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000008247 solid mixture Substances 0.000 description 1
- 230000000392 somatic effect Effects 0.000 description 1
- 235000011076 sorbitan monostearate Nutrition 0.000 description 1
- 239000001587 sorbitan monostearate Substances 0.000 description 1
- 229940035048 sorbitan monostearate Drugs 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 229960002920 sorbitol Drugs 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 208000010110 spontaneous platelet aggregation Diseases 0.000 description 1
- 238000012289 standard assay Methods 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 229940127230 sympathomimetic drug Drugs 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- XLLBNQDIURTZIE-UHFFFAOYSA-N tert-butyl 4-[2-[2-[(7-oxo-3-propan-2-yl-2,4-dihydropyrazolo[4,3-d]pyrimidin-5-yl)methyl]phenoxy]acetyl]piperazine-1-carboxylate Chemical compound CC(C)C1=NNC(C(N2)=O)=C1N=C2CC1=CC=CC=C1OCC(=O)N1CCN(C(=O)OC(C)(C)C)CC1 XLLBNQDIURTZIE-UHFFFAOYSA-N 0.000 description 1
- MHXBHWLGRWOABW-UHFFFAOYSA-N tetradecyl octadecanoate Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCCCCCCCCCCCCCC MHXBHWLGRWOABW-UHFFFAOYSA-N 0.000 description 1
- 210000001541 thymus gland Anatomy 0.000 description 1
- 239000003749 thyromimetic agent Substances 0.000 description 1
- 229960003087 tioguanine Drugs 0.000 description 1
- 210000003014 totipotent stem cell Anatomy 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 230000005030 transcription termination Effects 0.000 description 1
- 238000001890 transfection Methods 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- CYRMSUTZVYGINF-UHFFFAOYSA-N trichlorofluoromethane Chemical compound FC(Cl)(Cl)Cl CYRMSUTZVYGINF-UHFFFAOYSA-N 0.000 description 1
- 229940029284 trichlorofluoromethane Drugs 0.000 description 1
- PHYFQTYBJUILEZ-IUPFWZBJSA-N triolein Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC(OC(=O)CCCCCCC\C=C/CCCCCCCC)COC(=O)CCCCCCC\C=C/CCCCCCCC PHYFQTYBJUILEZ-IUPFWZBJSA-N 0.000 description 1
- 229940117972 triolein Drugs 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 230000002485 urinary effect Effects 0.000 description 1
- 238000012795 verification Methods 0.000 description 1
- 108700026220 vif Genes Proteins 0.000 description 1
- 238000010451 viral insertion Methods 0.000 description 1
- 238000012800 visualization Methods 0.000 description 1
- 230000004584 weight gain Effects 0.000 description 1
- 230000004580 weight loss Effects 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- 239000003871 white petrolatum Substances 0.000 description 1
- 239000000230 xanthan gum Substances 0.000 description 1
- 229920001285 xanthan gum Polymers 0.000 description 1
- 229940082509 xanthan gum Drugs 0.000 description 1
- 235000010493 xanthan gum Nutrition 0.000 description 1
- 229940075420 xanthine Drugs 0.000 description 1
- 239000000811 xylitol Substances 0.000 description 1
- 235000010447 xylitol Nutrition 0.000 description 1
- HEBKCHPVOIAQTA-SCDXWVJYSA-N xylitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)CO HEBKCHPVOIAQTA-SCDXWVJYSA-N 0.000 description 1
- 229960002675 xylitol Drugs 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
- C12N15/1137—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K67/00—Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
- A01K67/027—New or modified breeds of vertebrates
- A01K67/0275—Genetically modified vertebrates, e.g. transgenic
- A01K67/0276—Knock-out vertebrates
-
- 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
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
- A61P19/02—Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/24—Antidepressants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/04—Anorexiants; Antiobesity agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/08—Drugs for disorders of the metabolism for glucose homeostasis
- A61P3/10—Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
- A61P9/10—Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
- A61P9/12—Antihypertensives
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/8509—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0603—Embryonic cells ; Embryoid bodies
- C12N5/0606—Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/14—Hydrolases (3)
- C12N9/16—Hydrolases (3) acting on ester bonds (3.1)
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2217/00—Genetically modified animals
- A01K2217/07—Animals genetically altered by homologous recombination
- A01K2217/072—Animals genetically altered by homologous recombination maintaining or altering function, i.e. knock in
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2217/00—Genetically modified animals
- A01K2217/07—Animals genetically altered by homologous recombination
- A01K2217/075—Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2227/00—Animals characterised by species
- A01K2227/10—Mammal
- A01K2227/105—Murine
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2267/00—Animals characterised by purpose
- A01K2267/03—Animal model, e.g. for test or diseases
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2510/00—Genetically modified cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2517/00—Cells related to new breeds of animals
Definitions
- the present invention provides methods to decrease body weight and/or body fat in the treatment, for example, of overweight or obese patients, and methods to treat eating disorders (e.g., binge eating disorder and bulimia), by administering a phosphodiesterase 9 (PDE9) inhibitor.
- PDE9 phosphodiesterase 9
- the invention also features genetically- modified mammalian cells, and genetically-modified mice, with a disruption of the PDE9 gene.
- Cyclic nucleotide phosphodiesterases catalyze the hydrolysis of cyclic nucleotides, such as the second messengers cAMP (cyclic adenosine 3'5'- monophosphate) and cGMP (cyclic guanine 3'5'-monophosphate).
- PDEs play a pivotal regulatory role in a wide variety of signal transduction pathways (Beavo, Physiol. Rev. 75: 725-748, 1995).
- PDEs mediate processes involved in vision (McLaughlin et al., Nat. Genet. 4: 130-134, 1993), olfaction (Yan et al., Proc. Natl. Acad. Sci. USA 92: 9677-81 , 1995), platelet aggregation (Dickinson et al. Biochem. J. 323: 371-377, 1997), aldosterone synthesis (MacFarland et al., J. Biol. Chem.
- PDEs form a superfamily of enzymes that are subdivided into 11 major gene families (Beavo, Physiol. Rev. 75: 725-748, 1995; Beavo et al., Mol. Pharmacol.
- Each PDE gene family encodes a phosphodiesterase distinguished functionally by unique enzymatic characteristics and pharmacological profiles. In addition, each family exhibits distinct tissue, cell, and subcellular expression patterns (Beavo et al., Mol. Pharmacol. 46: 399-405, 1994; Soderling et al., Proc. Natl. Acad. Sci. USA 95: 8991-8996, 1998; Fisher et al., Biochem. Biophys. Res. Commun. 246: 570-577, 1998; Hayashi et al., Biochem.
- PDE9 is present in a variety of human tissues, including testes, brain, small intestine, skeletal muscle, heart, lung, thymus, and spleen. PDE9 inhibitors have bee reported as useful to treat cardiovascular disorders (WO 03/037899), and insulin resistance syndrome, hypertension, and/or type 2 diabetes (WO 03/037432).
- the invention features a method of treating an animal to reduce body fat comprising administering to an animal in need thereof a therapeutically effective amount of a PDE9 inhibitor.
- the animal is a human or companion animal (e.g., dog, cat, horse) and is overweight, more preferably, the animal is obese.
- the animal is a food stock animal (e.g., chicken, cattle, pig) and such treatment is rendered to produce leaner meat.
- the PDE9 inhibitor is a PDE9 selective inhibitor or the PDE9 inhibitor is administered orally.
- the invention features a method of treating an animal for an eating disorder comprising administering to an animal in need thereof a therapeutically effective amount of a PDE9 inhibitor.
- the eating disorder is binge eating disorder or bulimia
- the PDE9 inhibitor is a PDE9 selective inhibitor
- the PDE9 inhibitor is administered orally.
- the invention features a method of treating an animal for metabolic syndrome comprising administering to an animal in, need thereof a therapeutically effective amount of a PDE9 inhibitor.
- the PDE9 inhibitor is a PDE9 selective inhibitor, or the PDE9 inhibitor is administered orally.
- the invention also features a genetically-modified mouse, wherein the mouse is homozygous for disruption of the PDE9 gene and wherein the mouse, following a six week high fat diet, exhibits reduced body weight or reduced fat mass in an adipose depot, as compared to a wild type mouse following a six week high fat diet.
- the mouse expresses an exogenous reporter gene under the control of the regulatory sequences of the PDE9 gene or the mouse exhibits nondetectable PDE9 activity.
- the invention provides a cultured genetically-modified murine cell derived from the above-described mouse.
- the invention provides a method for producing the above-described mouse comprising: (a) introducing a DNA sequence into a mouse ES cell, wherein the DNA sequence comprises a PDE9 gene targeting construct, which, upon recombination with the PDE9 gene, disrupts the PDE9 gene; (b) selecting a mouse ES cell whose genome comprises a disruption of the PDE9 gene; (c) introducing an ES cell selected in step (b) into a mouse blastocyst or morulae; (d) transplanting the blastocyst or morulae of step (c) into a pseudopregnant mouse; (e) developing the transferred blastocyst or morulae to term to produce a chimeric mouse; and (f) mating sexually mature chimeric mice and mice heterozygous for the PDE9 disruption to obtain a mouse homozygous for the PDE9 gene disruption; wherein the mouse, following a six week high fat diet, exhibits reduced body weight or reduced
- the invention also features a genetically-modified cultured mammalian cell, wherein the cell is homozygous for disruption of the PDE9 gene and wherein the cell, or a progeny cell derived from the cell, exhibits nondetectable PDE9 polypeptide activity wherein the cell or progeny cell would exhibit PDE9 polypeptide activity absent the homozygous disruption.
- the cell is an embryonic stem (ES) cell, more preferably, the cell is a murine ES cell or a human ES cell.
- the invention provides an isolated nucleic acid molecule comprising a PDE11 gene targeting construct, wherein, upon recombination with the PDE9 gene, the construct disrupts the PDE9 gene.
- PDE9 inhibitor an agent that reduces or attenuates the biological activity of the PDE9 polypeptide.
- Such agents may include proteins, such as anti-PDE9 antibodies, nucleic acids, e.g., PDE9 antisense or RNA interference (RNAi) nucleic acids, amino acids, peptides, carbohydrates, small molecules (organic or inorganic), or any other compound or composition which decreases the activity of a PDE9 polypeptide either by effectively reducing the amount of PDE9 present in a cell, or by decreasing the enzymatic activity of the PDE9 polypeptide.
- RNAi RNA interference
- Compounds that are PDE9 inhibitors include all solvates, hydrates, pharmaceutically acceptable salts, tautomers, stereoisomers, and prodrugs of the compounds.
- a small molecule PDE9 inhibitor used in the present invention has an IC 50 of less that 10 ⁇ M, more preferably, less than 1 ⁇ M, and, even more preferably, less than 0.1 ⁇ M.
- Any PDE9 inhibitor used in the present invention is preferably also selective against some or all other PDEs, preferably, against PDE1A, PDE1 B, PDE1C, PDE2, PDE3A, PDE3B, PDE4A, PDE4B, PDE4C, PDE4D, PDE5, PDE6, PDE7A, PDE7B, PDE8A, PDE8B, PDE10, and/or PDE11.
- a “selective" PDE9 inhibitor is meant an agent that inhibits PDE9 activity with an IC 50 at least 10-fold less, preferably, at least 100-fold less, than the IC 50 for inhibition of one or more other PDEs.
- agents are combined with a pharmaceutically acceptable delivery vehicle or carrier.
- An antisense oligonucleotide directed to the PDE9 gene or mRNA to inhibit its expression is made according to standard techniques (see, e.g., Agrawal et al. Methods in Molecular Biology: Protocols for Oligonucleotides and Analogs, Vol. 20, 1993).
- RNA molecule that functions to reduce the production of PDE9 enzyme in a cell can be produced according to standard techniques known to those skilled in the art (see, e.g., Hannon, Nature 418: 244-251 , 2002; Shi, Trends in Genetics 19: 9-12, 2003; Shuey et al., Drug Discovery Today 7: 1040-1046, 2002).
- Examples of PDE9 inhibitors are provided herein and in WO 03/037899, in WO 03/037432, and in U.S. Provisional Patent Appl. No. 60/466,639, filed April 30, 2003, incorporated herein by reference.
- Decreased PDE9 activity means a manipulated decrease in the polypeptide activity of the PDE9 enzyme as a result of genetic disruption or manipulation of the PDE9 gene function that causes a reduction in the level of functional PDE9 polypeptide in a cell, or as the result of administration of a pharmacological agent that inhibits PDE9 activity.
- pharmaceutically acceptable indicates that the designated carrier, vehicle, diluent, excipient(s), and/or salt is generally chemically and/or physically compatible with the other ingredients comprising the formulation, and physiologically compatible with the recipient thereof.
- prodrug refers to a compound that is a drug precursor which, following administration, releases the drug in vivo via a chemical or physiological process (e.g., upon being brought to physiological pH or through enzyme activity).
- a discussion of the synthesis and use of prodrugs is provided by Higuchi and Stella, Prodrugs as Novel Delivery Systems, vol. 14 of the ACS Symposium Series, and Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
- salts and “pharmaceutically acceptable salts” refer to organic and inorganic salts of a compound, a stereoisomer of the compound, or a prodrug of the compound.
- BMI body mass index
- Obesity is typically defined as a BMI of 30 or greater (see, e.g., National Heart, Lung, and Blood Institute, Clinical Guidelines on the Identification, Evaluation, and Treatment of Overweight and Obesity in Adults, The Evidence Report, Washington, DC: U.S. Department of Health and Human Services, NIH publication no. 98-4083,1998).
- BMI National Heart, Lung, and Blood Institute
- Clinical Guidelines on the Identification, Evaluation, and Treatment of Overweight and Obesity in Adults The Evidence Report, Washington, DC: U.S. Department of Health and Human Services, NIH publication no. 98-4083, 1998.
- BMI body fat
- NIH publication no. 98-4083 International Health and Human Services
- a “high fat diet” as administered to a genetically-modified or wild type mouse, is meant a diet composed of at least 45% kcal fat, and, preferably, at least 58% fat.
- Exemplary diets include the Surwit diet (Surwit et al., Metabolism 47: 1354-1359; Surwit et al., Metabolism 47: 1089-1096, 1998; Surwit et al., J. Biol. Chem.
- High blood pressure ⁇ 60/90 mmHg
- Hyperlipidemia triglyceride concentration ⁇ 50 mg/dl (1.695 mmol/l) and/or HDL cholesterol ⁇ 35 mg/dl (0.9 mmol/l) in men and ⁇ 39 mg/dl (1.0 mmol/l) in women;
- Microalbuminuria urinary albumin excretion rate ⁇ >0 g/min or an albumin-to- creatinine ratio ⁇ 0 mg/kg.
- therapeutically effective is meant resulting in a decrease in body fat.
- a “disrupted PDE9 gene” refers to a PDE9 gene that is genetically-modified such that the cellular activity of the PDE9 polypeptide encoded by the disrupted gene is decreased or, preferably, eliminated in cells that normally express a wild type version of the PDE9 gene.
- the genetic modification When the genetic modification effectively eliminates all wild type copies of the PDE9 gene in a cell (e.g., the genetically-modified, non-human mammal or animal cell is homozygous for the PDE9 gene disruption or the only wild type copy of the PDE9 gene originally present is now disrupted), the genetic modification results in a reduction in PDE9 polypeptide activity as compared to a control cell that expresses the wild type PDE9 gene.
- PDE9 polypeptide activity results from either reduced PDE9 gene expression (i.e., PDE9 mRNA levels are effectively reduced resulting in reduced levels of PDE9 polypeptide) and/or because the disrupted PDE9 gene encodes a mutated polypeptide with altered, e.g., reduced, function as compared to a wild type PDE9 polypeptide.
- the activity of PDE9 polypeptide in the genetically-modified, non-human mammal or animal cell is reduced to 50% or less of wild type levels, more preferably, to 25% or less, and, even more preferably, to 10% or less of wild type levels.
- the homozygous PDE9 gene disruption results in non-detectable PDE9 activity in cells of a type that demonstrate wild type PDE9 activity.
- a "genetically-modified, non-human mammal" containing a disrupted PDE9 gene refers to a non-human mammal created by genetic engineering to contain a disrupted PDE9 gene, as well as a progeny of such non-human mammal that inherits the disrupted PDE9 gene.
- a genetically-modified non-human mammal may be produced, for example, by creating a blastocyst or embryo carrying the desired genetic modification and then implanting the blastocyst or embryo in a foster mother for in utero development.
- the genetically-modified blastocyst or embryo can be made, in the case of mice, by implanting a genetically-modified embryonic stem (ES) cell into a mouse blastocyst or by aggregating ES cells with tetraploid embryos.
- ES embryonic stem
- various species of genetically-modified embryos can be obtained by nuclear transfer.
- the donor cell is a somatic cell or a pluripotent stem cell, and it is engineered to contain the desired genetic modification that disrupts the PDE9 gene.
- the nucleus of this cell is then transferred into a fertilized or parthenogenetic oocyte that is enucleated; the resultant embryo is reconstituted and developed into a blastocyst.
- a genetically-modified blastocyst produced by either of the above methods is then implanted into a foster mother according to standard methods well known to those skilled in the art.
- a "genetically- modified, non-human mammal” includes all progeny of the non-human mammals created by the methods described above, provided that the progeny inherit at least one copy of the genetic modification that disrupts the PDE9 gene. It is preferred that all somatic cells and germline cells of the genetically-modified non-human mammal contain the modification.
- Preferred non-human mammals that are genetically- modified to contain a disrupted PDE9 gene include rodents, such as mice and rats, cats, dogs, rabbits, guinea pigs, hamsters, sheep, pigs, and ferrets.
- a "genetically-modified animal cell" containing a disrupted PDE9 gene refers to an animal cell (preferably a mammalian cell), including a human cell, created by genetic engineering to contain a disrupted PDE9 gene, as well as daughter cells and cells differentiated from a genetically-modified parent ES or stem cell, that inherit the disrupted PDE9 gene. These cells may be genetically-modified in culture according to any standard method known in the art.
- non-human mammalian cells may also be isolated from a genetically-modified, non-human mammal that contains a PDE9 gene disruption.
- the animal cells of the invention may be obtained from primary cell or tissue preparations as well as culture-adapted, tumorigenic, or transformed cell lines.
- These cells and cell lines are derived, for example, from endothelial cells, epithelial cells, islets, neurons and other neural tissue-derived cells, mesothelial cells, osteocytes, lymphocytes, chondrocytes, hematopoietic cells, immune cells, cells of the major glands or organs (e.g., testicle, liver, lung, heart, stomach, pancreas, kidney, and skin), muscle cells (including cells from skeletal muscle, smooth muscle, and cardiac muscle), exocrine or endocrine cells, fibroblasts, and embryonic and other totipotent or pluripotent stem cells (e.g., ES cells, ES-like cells, embryonic germline cells, and other stem cells, such as progenitor cells and tissue-derived stem cells).
- endothelial cells e.g., endothelial cells, epithelial cells, islets, neurons and other neural tissue-derived cells, mesothelial cells, osteocytes,
- the preferred genetically-modified cells are ES cells, more preferably, mouse or rat ES cells, and, most preferably, human ES cells, as well as cells differentiated from the genetically-modified ES cells.
- a non-human mammal or a animal cell that is "genetically-modified” is heterozygous or homozygous for a modification that is introduced into the non-human mammal or animal cell, or into a progenitor non-human mammal or animal cell, by genetic engineering.
- the standard methods of genetic engineering that are available for introducing the modification include homologous recombination, viral vector gene trapping, irradiation, chemical mutagenesis, and the transgenic expression of a nucleotide sequence encoding antisense RNA alone or in combination with catalytic ribozymes.
- Preferred methods for genetic modification to disrupt a gene are those which modify an endogenous gene by inserting a "foreign nucleic acid sequence" into the gene locus, e.g., by homologous recombination or viral vector gene trapping.
- a "foreign nucleic acid sequence” is an exogenous sequence that is non-naturally occurring in the gene.
- This insertion of foreign DNA can occur within any region of the PDE9 gene, e.g., in an enhancer, promoter, regulator region, noncoding region, coding region, intron, or exon.
- the most preferred method of genetic engineering for gene disruption is homologous recombination, in which the foreign nucleic acid sequence is inserted in a targeted manner either alone or in combination with a deletion of a portion of the endogenous gene sequence.
- "Homozygosity" when referring to PDE9 gene disruption in a non-human mammal or an animal cell means a non-human mammal or animal cell having disruption of all alleles of the PDE9 gene.
- the PDE9 gene sequences of each of these disrupted alleles need not be identical.
- a non-human mammal may be homozygous for PDE9 disruption wherein one allele of PDE9 is disrupted as a result of deletion of one region of the gene sequence and the other allele is disrupted as a result of deletion of another region of the gene sequence.
- ES cell or an "ES-like cell” means a pluripotent stem cell derived from an embryo, from a primordial germ cell, or from a teratocarcinoma, that is capable of indefinite self-renewal as well as differentiation into cell types that are representative of all three embryonic germ layers.
- “Microarray” means an arrangement of distinct polynucleotides or polypeptides on a substrate, as more fully described herein.
- Wild type when referring to a non-human mammal or an animal cell, means a non-human mammal or an animal cell, as the case may be, that does not comprise a disrupted PDE9 gene.
- wild type in a comparison of a particular characteristic of a non-human mammal of this invention to that characteristic in a wild type mammal, the term wild type refers to non-human mammal that does not comprise a disrupted PDE9 gene (i.e., a mammal whose PDE9 gene is wild type).
- a wild type non-human mammal is substantially similar, and, more preferably, substantially identical, to a non-human mammal of the invention, except for the non- disruption or disruption of the PDE9 gene, respectively.
- wild type refers to an animal cell that does not comprise a disrupted PDE9 gene (i.e., a cell whose PDE9 gene is wild type).
- a wild type animal cell is substantially similar, and, more preferably, substantially identical, to an animal cell of the invention, except for the non-disruption or disruption of the PDE9 gene, respectively.
- nucleic acids and polypeptides are found in standard textbooks of molecular biology, protein science, and immunology (see, e.g., Davis et al., Basic Methods in Molecular Biology, Elsevir Sciences Publishing, Inc., New York, NY, 1986; Hames et al., Nucleic Acid Hybridization, IL Press, 1985; Molecular
- FIG. 1 is a schematic of the targeting construct used to disrupt the PDE9 gene.
- a portion of the genomic sequence of each homology arm is shown as SEQ ID NO: 1 and SEQ ID NO: 2.
- Fig. 2 shows the cDNA sequence for a murine PDE9 (SEQ ID NO: 3).
- the underlined sequence, base pairs 142-175 was deleted and replaced with LacZ-Neo.
- FIG. 3 is a line graph detailing the body weight change in wild type (WT) and genetically-modified mice homozygous for disruption of the PDE9 gene (PDE9 knockout (KO) mice) during the course of a six week high fat diet.
- Fig. 4A male
- Fig. 4B female
- SC subcutaneous
- TBW total body weight
- Fig. 5 is a bar graph comparing the body weight of female WT and PDE9 KO mice following a six week control chow diet.
- Fig. 6A baseline
- Fig. 6A baseline
- 6B post-six week chow diet are bar graphs showing the mass of adipose depots in female WT and PDE9 KO mice. (Ing - inguinal subcutaneous; Gon - gonadal; RP - retroperitoneal; Mes - mesenteric) -13-
- PDE9 inhibitors are known to those skilled in the art and may be determined by standard assays known to those in the art, such as in WO 03/037899 and WO 03/037432.
- the PDE9 inhibitors used in the methods of the invention include those disclosed in WO 03/037899 and WO 03/037432, as well as in U.S. Provisional Appl. No. 60/466,639, filed April 30, 2003, incorporated hereinbefore by reference.
- Compound A 3-isopropyl-5-[2-(2-morpholin-4-yl-ethoxy)-benzyl]-1 ,6-dihydro-pyrazolo[4,3- d] pyrimidin-7-one (hereinafter referred to as "Compound A"); 1- ⁇ [2-(3-isopropyl-7-oxo-6,7-dihydro-1 H-pyrazolo[4,3-d]pyrimidin-5-ylmethyl)- phenoxy]-acetyl ⁇ -pyrrolidine-2-carboxylic acid; 3-isopropyl-5-[2-(2-oxo-2-piperazin-1-yl-ethoxy)-benzyl]-1 ,6-dihydro- pyrazolo[4,3-d]pyrimidin-7-one trifluoro acetate; 3-isopropyl-5-[2-(2-morpholin-4-yl-2-oxo-ethoxy)-benzyl]-1 ,6-dihydro
- Fig. 7 is a line graph detailing the time course for body weight gain in female ob/ob mice in Control, Compound A-treated (100 mg/kg/day), and Darglitazone- treated groups.
- Fig. 8A is a bar graph showing the Compound A dose effect on body weight at Days 2 and 4 in female ob/ob mice.
- Fig. 8B is a bar graph showing the Compound A dose effect on food consumption at Days 2 and 4 in female ob/ob mice.
- Fig. 9 is a bar graph comparing the time course for food consumption between Control, Compound A-treated (100 mg/kg/day), and Darglitazone-treated female ob/ob mice.
- Fig. 8A is a bar graph showing the Compound A dose effect on body weight at Days 2 and 4 in female ob/ob mice.
- Fig. 8B is a bar graph showing the Compound A dose effect on food consumption at Days 2 and 4 in female ob/ob mice.
- Fig. 10 is a line graph comparing plasma glucose in Control, Compound A- treated (100 mg/kg/day), and Darglitazone-treated female ob/ob mice.
- Fig. 11 is a line graph showing plasma triglycerides in Control and Compound A-treated (50 and 100 mg/kg/day) female ob/ob mice at Days 1 , 2, and 4.
- Fig. 12 is a bar graph comparing plasma fructosamine in Control, Compound A (100 mg/kg/day), and Darglitazone-treated female ob/ob mice at Day 16.
- the present invention is directed to methods to decrease body weight and/or body fat in an animal, e.g., in the treatment of overweight or obese patients (e.g., humans or companion animals), or as a means to produce leaner meat in food stock animals (e.g., cattle, chickens, pigs), and methods to treat eating disorders (e.g., binge eating disorder and bulimia) in patients in need thereof by administering a PDE9 inhibitor.
- the invention also features biological tools to further study PDE9 function, i.e., genetically-modified mice and animal cells having a PDE9 gene disruption.
- a PDE9 inhibitor reduces weight gain in the ob/ob mouse model of obesity
- PDE9 knockout mice are relatively resistant to developing increased body weight and increased adiposity subsequent to exposure to a high fat diet.
- Both Examples demonstrate that causing a decrease in PDE9 activity is an effective method to reduce body weight and/or body fat, and can be used, e.g., to treat animal patients that are overweight, obese, or suffer from an eating disorder, and can be used in animal food stock species to produce leaner meat.
- An agent identified as a PDE9 inhibitor is administered in a dose sufficient to reduce body weight or body fat, e.g., by reducing the mass of one or more adipose depots.
- Such therapeutically effective amounts will be determined using routine optimization techniques that are dependent on, for example, the condition of the patient or animal, the route of administration, the formulation, the judgment of the practitioner, and other factors evident to those skilled in the art in light of this disclosure.
- the PDE9 inhibitors suitable for use in accordance with the present invention can be administered alone but, in human therapy, will generally be administered in admixture with a suitable pharmaceutical excipient diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
- a suitable pharmaceutical excipient diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
- the PDE9 suitable for use in accordance with the present invention or salts or solvates thereof can be administered orally, buccally or sublingually in the form of tablets, capsules (including soft gel capsules), multi- particulate, gels, films, ovules, elixirs, solutions or suspensions, which may contain flavoring or coloring agents, for immediate-, delayed-, modified-, sustained-, dual-, controlled-release or pulsatile delivery applications.
- Such compounds may also be administered via fast dispersing or fast dissolving dosages forms or in the form of a high energy dispersion or as coated particles.
- Suitable pharmaceutical formulations may be in coated or un-coated form as desired.
- Such solid pharmaceutical compositions for example, tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate, glycine and starch (preferably corn, potato or tapioca starch), disintegrants such as sodium starch glycollate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethyl cellulose (HPMC), hydroxypropylcellulose (HPC), hydroxypropyl methylcellulose acetate succinate (HPMCAS), sucrose, gelatin and acacia.
- excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate, gly
- lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.
- Solid compositions of a similar type may also be employed as fillers in gelatin capsules or HPMC capsules.
- Preferred excipients in this regard include lactose, starch, a cellulose, milk sugar or high molecular weight polyethylene glycols.
- the PDE9 inhibitor compounds may be combined with various sweetening or flavouring agents, colouring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
- Modified release and pulsatile release dosage forms may contain excipients such as those detailed for immediate release dosage forms together with additional excipients that act as release rate modifiers, these being coated on and/or included in the body of the device.
- Release rate modifiers include, but are not exclusively limited to, HPMC, HPMCAS, methyl cellulose, sodium carboxymethylcellulose, ethyl cellulose, cellulose acetate, polyethylene oxide, Xanthan gum, Carbomer, ammonio methacrylate copolymer, hydrogenated castor oil, carnauba wax, paraffin wax, cellulose acetate phthalate, hydroxypropylmethyl cellulose phthalate, methacrylic acid copolymer and mixtures thereof.
- Modified release and pulsatile release dosage forms may contain one or a combination of release rate modifying excipients.
- Release rate modifying excipients maybe present both within the dosage form, i.e., within the matrix, and/or on the dosage form, i.e., upon the surface or coating.
- Fast dispersing or dissolving dosage formulations may contain the following ingredients: aspartame, acesulfame potassium, citric acid, croscarmellose sodium, crospovidone, diascorbic acid, ethyl acrylate, ethyl cellulose, gelatin, hydroxypropylmethyl cellulose, magnesium stearate, mannitol, methyl methacrylate, mint flavouring, polyethylene glycol, fumed silica, silicon dioxide, sodium starch glycolate, sodium stearyl fumarate, sorbitol, xylitol.
- FDDFs dispersing or dissolving as used herein to describe FDDFs are dependent upon the solubility of the drug substance used i.e., in cases where the drug substance is insoluble, a fast dispersing dosage form can be prepared, and, in cases where the drug substance is soluble, a fast dissolving dosage form can be prepared.
- the PDE9 inhibitors suitable for use in accordance with the present invention can also be administered parenterally, for example, intracavernosally, intravenously, intra-arterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally, intrasternally, intracranially, intramuscularly or subcutaneously, or they may be administered by infusion or needle-free techniques.
- a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood.
- the aqueous solutions should be suitably buffered (preferably to a pH of from about 3 to 9), if necessary.
- the preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well-known to those skilled in the art.
- the daily dosage level of the PDE9 inhibitors for use in the present invention will usually be from 1 to 500 mg (in single or divided doses). A preferred dosage range is about 1 mg to about 100 mg. The dosage may by via single dose, divided daily dose, or multiple daily dose.
- tablets or capsules of the PDE9 inhibitors suitable for use in accordance with the present invention may contain from 1 mg to 250 mg of active compound for administration singly or two or more at a time, as appropriate.
- Preferred tablets or capsules will contain about 1 mg to about 50 mg of active compound for administration singly or two or more at a time, as appropriate.
- the physician in any event will determine the actual dosage which will be most suitable for any individual patient and it will vary with the age, weight and response of the particular patient.
- PDE9 inhibitors suitable for use in accordance with the present invention can also be administered intranasally or by inhalation and are conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurized container, pump, spray or nebuliser with the use of a suitable propellant, e.g.
- the dosage unit may be determined by providing a valve to deliver a metered amount.
- the pressurised container, pump, spray or nebuliser may contain a solution or suspension of the active compound, e.g.
- Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of a compound of the invention and a suitable powder base such as lactose or starch. Aerosol or dry powder formulations are preferably arranged so that each metered dose or "puff' contains from 1 to 50 mg of a PDE9 inhibitor for delivery to the animal to be treated.
- the overall daily dose with an aerosol will be in the range of from 1 to 50 mg which may be administered in a single dose or, more usually, in divided doses throughout the day.
- the PDE9 inhibitors suitable for use in accordance with the present invention may also be formulated for delivery via an atomiser.
- Formulations for atomiser devices may contain the following ingredients as solubilisers, emulsifiers or suspending agents: water, ethanol, glycerol, propylene glycol, low molecular weight polyethylene glycols, sodium chloride, fluorocarbons, polyethylene glycol ethers, sorbitan trioleate, oleic acid.
- the PDE9 inhibitors suitable for use in accordance with the present invention can be administered in the form of a suppository or pessary, or they may be applied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder.
- the PDE9 inhibitors suitable for use in accordance with the present invention may also be dermally or transdermally administered, for example, by the use of a skin patch. They may also be administered by the pulmonary or rectal routes. The PDE9 inhibitors may also be administered by the ocular route.
- the compounds can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum.
- the PDE9 inhibitors suitable for use in accordance with the present invention can be formulated as a suitable ointment containing the active ingredient or agent suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water.
- Suitable lotion or cream suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
- the PDE9 inhibitors suitable for use in accordance with the present invention may also be used in combination with a cyclodextrin. Cyclodextrins are known to form inclusion and non-inclusion complexes with drug molecules.
- Formation of a drug-cyclodextrin complex may modify the solubility, dissolution rate, bioavailability and/or stability property of a drug molecule.
- Drug-cyclodextrin complexes are generally useful for most dosage forms and administration routes.
- the cyclodextrin may be used as an auxiliary additive, e.g. as a carrier, diluent or solubiliser.
- Alpha-, beta- and gamma- cyclodextrins are some of the most commonly used and suitable examples are described in WO 91/11172, WO 94/02518 and WO 98/55148.
- oral administration is the preferred route, being the most convenient.
- the drug may be administered parenterally, sublingually, or buccally.
- a PDE9 inhibitor is administered as a suitably acceptable formulation in accordance with normal veterinary practice and the veterinary surgeon will determine the dosing regimen and route of administration which will be most appropriate for a particular animal.
- Such animals include companion animals who are overweight, obese, or at risk of being overweight or obese.
- Other animals that may be treated according to the present invention are foodstock animals in order to obtain leaner meat than would be obtained absent treatment according to the present invention.
- Therapeutic efficacy of such PDE9 inhibitors can be determined in light of this disclosure by standard therapeutic procedures in cell cultures or experimental animals, e.g., for determining the ED 50 (the dose therapeutically effective in 50% of the population).
- the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
- the dosage may vary, for example, depending upon the formulation and the route of administration.
- the therapeutically effective dose can be estimated initially from cell culture assays.
- a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
- Levels in plasma may be measured, for example, by high performance liquid chromatography.
- the PDE9 inhibitors used in accordance with the present invention may also be used in conjunction with other pharmaceutical agents for the treatment of the diseases, conditions and/or disorders described herein. Therefore, methods of treatment that include administering PDE9 inhibitors in combination with other pharmaceutical agents are also provided.
- Suitable pharmaceutical agents that may be used in combination with the compounds of the present invention include anti- obesity agents such as ⁇ 3 adrenergic receptor agonists, apolipoprotein-B secretion/microsomal triglyceride transfer protein (apo-B/MTP) inhibitors, peptide YY 3- 36 and analogs thereof, MCR-4 agonists, cholecystokinin-A (CCK-A) agonists, monoamine reuptake inhibitors (e.g., sibutramine), sympathomimetic agents, cannabinoid receptor antagonists (e.g., rimonabant (SR-141,716A)), dopamine agonists (e.g., bromocriptine), melanocyte-stimulating hormone receptor analogs, 5HT2c agonists, melanin concentrating hormone antagonists, leptin (the OB protein), leptin analogs, leptin receptor agonists, galanin antagonists, lipase inhibitors
- anti-obesity agents including the preferred agents set forth hereinbelow, are well known, or will be readily apparent in light of the instant disclosure, to one of ordinary skill in the art.
- anti-obesity agents selected from the group consisting of orlistat, sibutramine, bromocriptine, ephedrine, leptin, pseudoephedrine, and peptide YY 3-36 (including analogs thereof).
- compounds of the present invention and combination therapies are administered in conjunction with exercise and a sensible diet.
- anti-obesity agents for use in the combinations, pharmaceutical compositions, and methods of the invention can be prepared using methods known to one of ordinary skill in the art, for example, sibutramine can be prepared, e.g., as described in U.S. Pat. No. 4,929,629; bromocriptine can be prepared, e.g., as described in U.S. Pat. Nos. 3,752,814 and 3,752,888; orlistat can be prepared, e.g., as described in U.S. Pat. Nos. 5,274,143; 5,420,305; 5,540,917; and 5,643,874; and PYY 3-36 (including analogs) can be prepared, e.g., as described in U.S. Patent Appl.
- treatment of a mammal with a therapeutically effective amount of a PDE9 inhibitor can include a single treatment or, preferably, can include a series of treatments.
- the genetically-modified, non-human mammals and genetically-modified animal cells, including human cells, of the invention are heterozygous or homozygous for a modification that disrupts the PDE9 gene.
- the animal cells may be derived by genetically engineering cells in culture, or, in the case of non-human mammalian cells, the cells may be isolated from genetically-modified, non-human mammals.
- the PDE9 gene locus may be disrupted using techniques for genetic modification known in the art, including chemical mutagenesis (Rinchik, Trends in Genetics 7: 15-21 , 1991 , Russell, Environmental & Molecular Mutagenesis 23 (Suppl. 24): 23-29, 1994), irradiation (Russell, supra), transgenic expression of PDE9 gene antisense RNA, either alone or in combination with a catalytic RNA ribozyme sequence (Luyckx et al., Proc. Natl. Acad. Sci.
- the disruption of the PDE9 gene by the insertion of a foreign nucleic acid sequence into the PDE9 gene locus.
- the foreign sequence is inserted by homologous recombination or by the insertion of a viral vector.
- the method of PDE9 gene disruption to create the genetically modified non-human mammals and animal cells of the invention is homologous recombination and includes a deletion of a portion of the endogenous PDE9 gene sequence.
- the integration of the foreign sequence disrupts the PDE9 gene through one or more of the following mechanisms: by interfering with the PDE9 gene transcription or translation process (e.g., by interfering with promoter recognition, or by introducing a transcription termination site or a translational stop codon into the PDE9 gene); or by distorting the PDE9 gene coding sequence such that it no longer encodes a PDE9 polypeptide with normal function (e.g., by inserting a foreign coding sequence into the PDE9 gene coding sequence, by introducing a frameshift mutation or amino acid(s) substitution, or, in the case of a double crossover event, by deleting a portion of the PDE9 gene coding sequence that is required for expression of a functional PDE9 protein).
- the foreign DNA sequence is introduced into the cell according to a standard method known in the art such as electroporation, calcium-phosphate precipitation, retroviral infection, microinjection, biolistics, liposome transfection, DEAE-dextran transfection, or transferrinfection , (see, e.g., Neumann et al., EMBO J. 1 : 841-845, 1982; Potter et al., Proc. Natl. Acad. Sci USA 81 : 7161-65, 1984; Chu et al., Nucleic Acids Res.
- a standard method known in the art such as electroporation, calcium-phosphate precipitation, retroviral infection, microinjection, biolistics, liposome transfection, DEAE-dextran transfection, or transferrinfection , (see, e.g., Neumann et al., EMBO J. 1 : 841-845, 1982; Potter et al., Proc. Natl. Acad
- the preferred method for introducing foreign DNA into a cell is electroporation.
- Homologous Recombination targets the PDE9 gene for disruption by introducing a PDE9 gene targeting vector into a cell containing a PDE9 gene.
- the ability of the vector to target the PDE9 gene for disruption stems from using a nucleotide sequence in the vector that is homologous, i.e., related, to the PDE9 gene.
- This homology region facilitates hybridization between the vector and the endogenous sequence of the PDE9 gene.
- the probability of a crossover event between the targeting vector and genomic sequences greatly increases. This crossover event results in the integration of the vector sequence into the PDE9 gene locus and the functional disruption of the PDE9 gene.
- General principles regarding the construction of vectors used for targeting are reviewed in Bradley et al. (Biotechnol. 10: 534, 1992).
- Two different types of vector can be used to insert DNA by homologous recombination: an insertion vector or a replacement vector.
- An insertion vector is circular DNA which contains a region of PDE9 gene homology with a double stranded break.
- the more preferred vector to create the genetically modified non-human mammals and animals cells of the invention by homologous recombination is a replacement vector, which is colinear rather than circular.
- Replacement vector integration into the PDE9 gene requires a double crossover event, i.e., crossing over at two sites of hybridization between the targeting vector and the PDE9 gene.
- This double crossover event results in the integration of a vector sequence that is sandwiched between the two sites of crossover into the PDE9 gene and the deletion of the corresponding endogenous PDE9 gene sequence that originally spanned between the two sites of crossover (see, e.g., Thomas and Capecchi et al., Cell 51 : 503-12, 1987; Mansour et al., Nature 336: 348-52, 1988; Mansour et al., Proc. Natl. Acad. Sci. USA 87: 7688-7692, 1990; and Mansour, GATA 7: 219- 227, 1990).
- a region of homology in a targeting vector used to create the genetically modified non-human mammals and animal cells of the invention is generally at least 100 nucleotides in length. Most preferably, the homology region is at least 1- 5 kilobases (kb) in length. Although there is no demonstrated minimum length or minimum degree of relatedness required for a homology region, targeting efficiency for homologous recombination generally corresponds with the length and the degree of relatedness between the targeting vector and the PDE9 gene locus. In the case where a replacement vector is used, and a portion of the endogenous PDE9 gene is deleted upon homologous recombination, an additional consideration is the size of the deleted portion of the endogenous PDE9 gene.
- cloning vectors may be used as vector backbones in the construction of the PDE9 gene targeting vectors of the present invention, including pBluescript-related plasmids (e.g., Bluescript KS+11 ), pQE70, pQE60, pQE-9, pBS, pD10, phagescript, phiX174, pBK Phagemid, pNH8A, pNH16a, pNH18Z, pNH46A, ptrc99a, pKK223-3, pKK233-3, pDR540, and pRIT5 PWLNEO, pSV2CAT, pXT1 , pSG (Stratagene), pSVK3, PBPV, PMSG, and pSVL, pBR322 and pBR322-based vectors, pMB9, pBR325, pKH47, p
- pBluescript-related plasmids e.g.,
- vectors are available from a variety of commercial sources (e.g., Boehringer Mannheim Biochemicals, Indianapolis, IN; Qiagen, Valencia, CA; Stratagene, La Jolla, CA; Promega, Madison, Wl; and New England Biolabs, Beverly, MA).
- any other vectors e.g. plasmids, viruses, or parts thereof, may be used as long as they are replicable and viable in the desired host.
- the vector may also comprise sequences which enable it to replicate in the host whose genome is to be modified. The use of such a vector can expand the interaction period during which recombination can occur, increasing the efficiency of targeting (see Molecular Biology, ed. Ausubel et al, Unit 9.16, Fig. 9.16.1).
- the specific host employed for propagating the targeting vectors of the present invention is not critical. Examples include E. coli K12 RR1 (Bolivar et al., Gene 2: 95, 1977), E co// K12 HB101 (ATCC No. 33694), E. coli MM21 (ATCC No. 336780), E. coli DH1 (ATCC No. 33849), E. coli strain DH5 ⁇ , and E. coli STBL2.
- hosts such as C. cerevisiae or ⁇ . subtilis can be used. The above-mentioned hosts are available commercially (e.g., Stratagene, La Jolla, CA; and Life Technologies, Rockville, MD).
- a PDE9 gene targeting construct is added to an above-described vector backbone.
- the PDE9 gene targeting constructs of the invention have at least one PDE9 gene homology region.
- a PDE9 genomic or cDNA sequence is used as a basis for producing PCR primers. These primers are used to amplify the desired region of the PDE9 sequence by high fidelity PCR amplification (Mattila et al., Nucleic Acids Res. 19: 4967, 1991; Eckert and Kunkel 1 : 17, 1991 ; and U.S. Pat. No. 4,683, 202).
- the genomic sequence is obtained from a genomic clone library or from a preparation of genomic DNA, preferably from the animal species that is to be targeted for PDE9 gene disruption, a PDE9 cDNA sequence can be used in making a PDE9 targeting vector (e.g., GenBank® NM008804 (murine) or GenBank® NM002606 (human)).
- a PDE9 targeting vector e.g., GenBank® NM008804 (murine) or GenBank® NM002606 (human)
- the targeting constructs of the invention also include an exogenous nucleotide sequence encoding a positive marker protein.
- the stable expression of a positive marker after vector integration confers an identifiable characteristic on the cell, ideally, without compromising cell viability.
- the marker gene is positioned between two flanking homology regions so that it integrates into the PDE9 gene following the ' double crossover event in a manner such that the marker gene is positioned for expression after integration.
- the positive marker protein is a selectable protein; the stable expression of such a protein in a cell confers a selectable phenotypic characteristic, i.e., the characteristic enhances the survival of the cell under otherwise lethal conditions.
- the selectable condition one can isolate cells that stably express the positive selectable marker-encoding vector sequence from other cells that have not successfully integrated the vector sequence on the basis of viability.
- positive selectable marker proteins examples include neo (G418 or kanomycin), hyg (hygromycin), hisD (histidinol), gpt (xanthine), ble (bleomycin), and hprt (hypoxanthine) (see, e.g., Capecchi and Thomas, U.S. Pat. No. 5,464,764, and Capecchi, Science 244: 1288-92, 1989).
- positive markers that may also be used as an alternative to a selectable marker include reporter proteins such as ⁇ - galactosidase, firefly luciferase, or green fluorescent protein (see, e.g., Current Protocols in Cytometry, Unit 9.5, and Current Protocols in Molecular Biology, Unit 9.6, John Wiley & Sons, New York, NY, 2000).
- reporter proteins such as ⁇ - galactosidase, firefly luciferase, or green fluorescent protein
- PDE9 gene locus versus random, non-homologous integration of vector sequence into any chromosomal position. Therefore, when using a replacement vector for homologous recombination to make the genetically modified non-human mammals and animal cells of the invention, it is also preferred to include a nucleotide sequence encoding a negative selectable marker protein. Expression of a negative selectable marker causes a cell expressing the marker to lose viability when exposed to a certain agent (i.e., the marker protein becomes lethal to the cell under certain selectable conditions).
- negative selectable markers examples include herpes simplex virus thymidine kinase (gancyclovir or 1 ,2-deoxy-2-fluoro-c/-d-arabinofuransyl-5-iodouracil), Hprt (6- thioguanine or 6-thioxanthine), and diphtheria toxin, ricin toxin, and cytosine deaminase (5-fluorocytosine).
- the nucleotide sequence encoding the negative selectable marker is positioned outside of the two homology regions of the replacement vector.
- cells will only integrate and stably express the negative selectable marker if integration occurs by random, non-homologous recombination; homologous recombination between the PDE9 gene and the two regions of homology in the targeting construct excludes the sequence encoding the negative selectable marker from integration.
- cells that have integrated the targeting vector by random, non- homologous recombination lose viability.
- a targeting construct used to make the genetically modified non-human mammals and animal cells of the invention because a series of positive and negative selection steps can be designed to more efficiently select only those cells that have undergone vector integration by homologous recombination, and, therefore, have a potentially disrupted PDE9 gene.
- positive-negative selection schemes, selectable markers, and targeting constructs are described, for example, in U.S. Pat. No. 5,464,764, WO 94/06908, U.S. Pat. No. 5,859,312, and Valancius and Smithies, Mol. Cell. Biol. 11: 1402, 1991.
- the targeting vector may be designed so that the marker coding sequence is operably linked to the endogenous PDE9 gene promoter upon vector integration. Expression of the marker is then driven by the PDE9 gene promoter in cells that normally express the PDE9 gene.
- each marker in the targeting construct of the vector may contain its own promoter that drives expression independent of the PDE9 gene promoter. This latter scheme has the advantage of allowing for expression of markers in cells that do not typically express the PDE9 gene (Smith and Berg, Cold Spring Harbor Symp. Quant. Biol. 49: 171 , 1984; Sedivy and Sharp, Proc. Natl. Acad. Sci.
- Exogenous promoters that can be used to drive marker gene expression include cell-specific or stage-specific promoters, constitutive promoters, and inducible or regulatable promoters.
- Non-limiting examples of these promoters include the herpes simplex thymidine kinase promoter, cytomegalovirus (CMV) promoter/enhancer, SV40 promoters, PGK promoter, PMC1-neo, metallothionein promoter, adenovirus late promoter, vaccinia virus 7.5K promoter, avian beta globin promoter, histone promoters (e.g., mouse histone H3-614), beta actin promoter, neuron-specific enolase, muscle actin promoter, and the cauliflower mosaic virus 35S promoter (see generally, Sambrook et al., Molecular Cloning, Vols.
- primers or genomic probes that are specific for the desired vector integration event can be used in combination with polymerase chain reaction (PCR) or Southern blot analysis to identify the presence of the desired vector integration into the PDE9 gene locus (Erlich et al., Science 252: 1643-51 , 1991; Zimmer and Gruss, Nature 338: 150, 1989; Mouellic et al., Proc. Natl.
- PCR polymerase chain reaction
- Gene Trapping Another method available for inserting a foreign nucleic acid sequence into the PDE9 gene locus to disrupt the PDE9 gene, based on the present description, is gene trapping. This method takes advantage of the cellular machinery present in all mammalian cells that splices exons into mRNA to insert a gene trap vector coding sequence into a gene in a random fashion. Once inserted, the gene trap vector creates a mutation that may disrupt the trapped PDE9 gene.
- Individual mutant cell lines containing a disrupted PDE9 gene are identified in a population of mutated cells using, for example, reverse transcription (RT) and PCR to identify a mutation in a PDE9 gene sequence.
- RT reverse transcription
- This process can be streamlined by pooling clones. For example, to find an individual clone containing a disrupted PDE9 gene, RT-PCR is performed using one primer anchored in the gene trap vector and the other primer located in the PDE9 gene sequence.
- a positive RT-PCR result indicates that the vector sequence is encoded in the PDE9 gene transcript, indicating that the PDE9 gene has been disrupted by a gene trap integration event (see, e.g., Sands et al., WO 98/14614, U.S. Pat. No. 6,080,576).
- Temporal, Spatial, and Inducible PDE9 Gene Disruptions In certain embodiments of the present invention, a functional disruption of the endogenous PDE9 gene occurs at specific developmental or cell cycle stages (temporal disruption) or in specific cell types (spatial disruption). In other embodiments, the PDE9 gene disruption is inducible when certain conditions are present.
- a recombinase excision system such as a Cre-Lox system, may be used to activate or inactivate the PDE9 gene at a specific developmental stage, in a particular tissue or cell type, or under particular environmental conditions.
- Cre-Lox technology methods utilizing Cre-Lox technology are carried out as described by Torres and Kuhn, Laboratory Protocols for Conditional Gene Targeting, Oxford University Press, 1997. Methodology similar to that described for the Cre-Lox system can also be employed utilizing the FLP-FRT system. Further guidance regarding the use of recombinase excision systems for conditionally disrupting genes by homologous recombination or viral insertion is provided, for example, in U.S. Pat. No. 5,626,159; U.S. Pat. No. 5,527,695; U.S. Pat. No. 5,434,066; WO 98/29533; U.S. Pat. No. 6,228,639; Orban et al., Proc. Nat.
- More than one recombinase system can be used to genetically modify a non-human mammal or animal cell of the present invention.
- a portion of the PDE9 gene coding region is replaced by a targeting construct comprising the PDE9 gene coding region flanked by loxP sites.
- Non-human mammals and animal cells carrying this genetic modification contain a functional, loxP-flanked PDE9 gene.
- the temporal, spatial, or inducible aspect of the PDE9 gene disruption is caused by the expression pattern of an additional transgene, a Cre recombinase transgene, that is expressed in the non-human mammal or animal cell under the control of the desired spatially-regulated, temporally-regulated, or inducible promoter, respectively.
- a Cre recombinase targets the loxP sites for recombination. Therefore, when Cre expression is activated, the LoxP sites undergo recombination to excise the sandwiched PDE9 gene coding sequence, resulting in a functional disruption of the PDE9 gene (Rajewski et al., J. Clin. Invest.
- a cell containing both a Cre recombinase transgene and loxP-flanked PDE9 gene can be generated through standard transgenic techniques or, in the case of genetically-modified, non-human mammals, by crossing genetically- modified, non-human mammals wherein one parent contains a loxP flanked PDE9 gene and the other contains a Cre recombinase transgene under the control of the desired promoter. Further guidance regarding the use of recombinase systems and specific promoters to temporally, spatially, or conditionally disrupt the PDE9 gene is found, for example, in Sauer, Meth. Enz.
- This system involves genetically modifying a cell to introduce a Tet promoter into the endogenous PDE9 gene regulatory element and a transgene expressing a tetracycline-controllable repressor (TetR).
- TetR tetracycline-controllable repressor
- the administration of tetracycline activates the TetR which, in turn, inhibits PDE9 gene expression and, therefore, disrupts the PDE9 gene (St-Onge et al., Nucleic Acids Res. 24: 3875-77, 1996; U.S. Patent No. 5,922,927).
- Genetically- modified animal cells of the invention include, but are not limited to, mammalian cells, including human cells, and avian cells. These cells may be derived from genetically engineering any animal cell line, such as culture-adapted, tumorigenic, or transformed cell lines, differentiated genetically-engineered ES cells, or they may be isolated from a genetically-modified, non-human mammal carrying the desired PDE9 genetic modification. The cells may be heterozygous or homozygous for the disrupted PDE9 gene.
- the method uses a scheme in which PDE9+/- targeted clones that express a selectable drug resistance marker are selected against a very high drug concentration; this selection favors cells that express two copies of the sequence encoding the drug resistance marker and are, therefore, homozygous for the PDE9 gene disruption (Mortensen et al., Mol. Cell. Biol. 12: 2391-95, 1992).
- genetically-modified animal cells can be obtained from genetically-modified PDE9-/- non-human mammals that are created by mating non-human mammals that are PDE9+/- in germline ' cells, as further discussed below.
- the PDE9 gene locus can be confirmed as the site of modification by PCR analysis according to standard PCR or Southern blotting methods known in the art (see, e.g., U.S. Pat. No. 4,683,202; and Erlich et al., Science 252: 1643, 1991). Further verification of the functional disruption of the PDE9 gene may also be made if PDE9 gene messenger RNA (mRNA) levels and/or PDE9 polypeptide levels are reduced in cells that normally express the PDE9 gene. Measures of PDE9 gene mRNA levels may be obtained by using RT-PCR, Northern blot analysis, or in situ hybridization.
- mRNA messenger RNA
- the quantification of PDE9 polypeptide levels produced by the cells can be made, for example, by standard immunoassay methods known in the art.
- immunoassays include, but are not limited to, competitive and non- competitive assay systems using techniques such as RIAs (radioimmunoassays), ELISAs (enzyme-linked immunosorbent assays), "sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitin reactions, immunodiffusion assays, in situ immunoassays (using, for example, colloidal gold, enzymatic, or radioisotope labels), Western blots, 2-dimensional gel analysis, precipitation reactions, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays.
- Preferred genetically-modified animal cells of the invention are embryonic stem (ES) cells and ES-like cells. These cells are derived from the preimplantation embryos and blastocysts of various species, such as mice (Evans et al., Nature 129:154-156, 1981 ; Martin, Proc. Natl. Acad. Sci., USA, 78: 7634-7638, 1981), pigs and sheep (Notanianni et al., J. Reprod. Fert. Suppl., 43: 255-260, 1991 ; Campbell et al., Nature 380: 64-68,1996) and primates, including humans (Thomson et al., U.S. Patent No.
- a sample of ES cells can be cultured indefinitely as stem cells, allowed to differentiate into a wide variety of different cell types within a single sample, or directed to differentiate into a specific cell type, such as macrophage-like cells, neuronal cells, cardiomyocytes, chondrocytes, adipocytes, smooth muscle cells, endothelial cells, skeletal muscle cells, keratinocytes, and hematopoietic cells, such as eosinophils, mast cells, erythroid progenitor cells, or megakaryocytes.
- Directed differentiation is accomplished by including specific growth factors or matrix components in the culture conditions, as further described, for example, in Keller et al., Curr. Opin.
- exemplary murine ES cell lines include AB-1 (McMahon and Bradley, Cell 62:1073-85, 1990), E14 (Hooper et al., Nature 326: 292-95, 1987), D3 (Doetschman et al., J. Embryol. Exp. Morph. 87: 27-45, 1985), CCE (Robertson et al, Nature 323: 445-48, 1986), RW4 (Genome Systems, St. Louis, MO), and DBA/1 lacJ (Roach et al., Exp. Cell Res. 221 : 520-25, 1995); an exemplary human ES cell line is H1.1 cells (Zwaka and Thomson, Nature Biotech. 21 : 319-321 ,
- Genetically-modified murine ES cells may be used to generate genetically- modified mice, according to published procedures (Robertson, 1987, Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, Ed. E. J. Robertson, Oxford: IRL Press, pp. 71-112, 1987; Zjilstra et al., Nature 342: 435- 438, 1989; and Schwartzberg et al., Science 246: 799-803, 1989). Following confirmation that the ES cells contain the desired functional disruption of the PDE9 gene, these ES cells are then injected into suitable blastocyst hosts for generation of chimeric mice according to methods known in the art (Capecchi, Trends Genet. 5: 70, 1989).
- mice employed in the present invention are not critical.
- blastocysts include those derived from C57BL6 mice, C57BL6 Albino mice, Swiss outbred mice, CFLP mice, and MFI mice.
- ES cells may be sandwiched between tetraploid embryos in aggregation wells (Nagy et al., Proc. Natl. Acad. Sci. USA 90: 8424-8428, 1993).
- the blastocysts or embryos containing the genetically-modified ES cells are then implanted in pseudopregnant female mice and allowed to develop in utero (Hogan et al., Manipulating the Mouse Embryo: A Laboratory Manual, Cold Spring Harbor Laboratory press, Cold Spring Harbor, NY 1988; and Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E.J. Robertson, ed., IRL Press, Washington, D.C., 1987).
- the offspring born to the foster mothers may be screened to identify those that are chimeric for the PDE9 gene disruption.
- offspring contain some cells that are derived from the genetically-modified donor ES cell as well as other cells derived from the original blastocyst.
- offspring may be screened initially for mosaic coat color, where a coat color selection strategy has been employed, to distinguish cells derived from the donor ES cell from the other cells of the blastocyst.
- DNA from tail tissue of the offspring can be used to identify mice containing the genetically-modified cells. The mating of chimeric mice that contain the PDE9 gene disruption in germ line cells produces progeny that possess the PDE9 gene disruption in all germ line cells and somatic cells.
- mice that are heterozygous for the PDE9 gene disruption can then be crossed to produce homozygotes (see, e.g., U.S. Pat. No. 5,557,032; and U.S. Pat. No. 5,532,158).
- An alternative to the above-described ES cell technology for transferring a genetic modification from a cell to a whole animal is to use nuclear transfer.
- This method can be employed to make other genetically-modified, non-human mammals besides mice, for example, sheep (McCreath et al., Nature 29: 1066-69, 2000; Campbell et al., Nature 389: 64-66, 1996; and Schnieke et al., Science 278: 2130-33, 1997) and calves (Cibelli et al., Science 280: 1256-58, 1998).
- somatic cells e.g., fibroblasts
- pluripotent stem cells e.g., ES-like cells
- a promoterless marker be used in the vector such that vector integration into the PDE9 gene results in expression of the marker under the control of the PDE9 gene promoter (Sedivy and Dutriaux, T.I.G. 15: 88-90, 1999; McCreath et al., Nature 29: 1066-69, 2000). Nuclei from donor cells which have the appropriate PDE9 gene disruption are then transferred to fertilized or parthenogenetic oocytes that are enucleated (Campbell et al., Nature 380: 64, 1996; Wilmut et al., Nature 385: 810, 1997).
- the present invention also encompasses the progeny of the genetically- modified, non-human mammals and genetically-modified animal cells. While the progeny are heterozygous or homozygous for the genetic modification that disrupts the PDE9 gene, they may not be genetically identical to the parent non- human mammals and animal cells due to mutations or environmental influences, besides that of the original genetic disruption of the PDE9 gene, that may occur in succeeding generations.
- the cells from a non-human genetically modified animal can be isolated from tissue or organs using techniques known to those of skill in the art.
- the genetically modified cells of the invention are immortalized.
- cells can be immortalized by genetically engineering the telomerase gene, an oncogene, e.g., mos or v-src, or an apoptosis-inhibiting gene, e.g., bcl-2, into the cells.
- cells can be immortalized by fusion with a hybridization partner utilizing techniques known to one of skill in the art.
- “Humanized” Non-human Mammals and Animal Cells The genetically-modified non-human mammals and animal cells (non- human) of the invention containing a disrupted endogenous PDE9 gene can be further modified to express the human PDE9 sequence (referred to herein as "humanized”).
- a preferred method for humanizing cells involves replacing the endogenous PDE9 sequence with nucleic acid sequence encoding the human PDE9 sequence (Jakobsson et al., Proc. Natl. Acad. Sci. USA 96: 7220-25, 1999) by homologous recombination.
- the vectors are similar to those traditionally used as targeting vectors with respect to the 5' and 3' homology arms and positive/negative selection schemes.
- the vectors also include sequence that, after recombination, either substitutes the human PDE9 coding sequence for the endogenous sequence, or effects base pair changes, exon substitutions, or codon substitutions that modify the endogenous sequence to encode the human PDE9.
- the human sequence can be the full length human cDNA sequence with a polyA tail attached at the 3' end for proper processing or the whole genomic sequence (Shiao et al., Transgenic Res. 8: 295-302, 1999). Further guidance regarding these methods of genetically modifying cells and non-human mammals to replace expression of an endogenous gene with its human counterpart is found, for example, in Sullivan et al., J. Biol. Chem. 272: 17972-17980, 1997, Reaume et al., J. Biol. Chem. 271 : 23380-23388, 1996, and Scott et al., U.S. Pat. No. 5,777,194).
- Another method for creating such "humanized” organisms is a two step process involving the disruption of the endogenous gene followed by the introduction of a transgene encoding the human sequence by pronuclear microinjection into the knock-out embryos.
- Uses for the Genetically-Modified Non-human Mammals and Animal Cells PDE9 function and therapeutic relevance can be further elucidated by additional investigation into the phenotype of PDE9-/- non-human mammals and animals cells of the invention, as illustrated, for example, in the Examples hereinbelow.
- the genetically-modified PDE9-/- non-human mammals and animal cells can be used to determine whether the PDE9 plays a role in causing or preventing symptoms or phenotypes to develop in certain models of disease, e.g., obesity, eating disorders, cardiovascular disorders, insulin resistance syndrome, hypertension, and/or type 2 diabetes. If a symptom or phenotype is different in a PDE9-/- non-human mammal or animal cell as compared to a wild type (PDE9+/+) or PDE9+/- non-human mammal or animal cell, then the PDE9 polypeptide plays a role in regulating functions associated with the symptom or phenotype.
- diseases or phenotypes e.g., obesity, eating disorders, cardiovascular disorders, insulin resistance syndrome, hypertension, and/or type 2 diabetes.
- Examples of testing that can be used to assess PDE9 function include comparing PDE9-/- mice to wild type mice in terms of body weight, body fat, blood pressure, glucose/insulin metabolism (e.g., glucose uptake in isolated tissues, alterations in the activity of glycogen metabolism enzymes, alterations in glycogen levels in liver or muscle, and/or alterations in body composition), and changes in the activity or phosphorylation state of components in the insulin signaling pathway.
- glucose/insulin metabolism e.g., glucose uptake in isolated tissues, alterations in the activity of glycogen metabolism enzymes, alterations in glycogen levels in liver or muscle, and/or alterations in body composition
- the genetically-modified PDE9-/- non-human mammals and animal cells of the invention are useful to characterize any other effects caused by the agent besides those known to result from the (ant)agonism of PDE9 (i.e., the non-human mammals and animal cells can be used as negative controls).
- the administration of the agent causes an effect in a PDE9+/+ non-human mammal or animal cell that is not known to be associated with PDE9 polypeptide activity
- the agent exerts this effect solely or primarily through modulation of PDE9 by administering the agent to a corresponding PDE9-/- non-human mammal or animal cell. If this effect is absent, or is significantly reduced, in the PDE9-/- non- human mammal or animal cell, then the effect is mediated, at least in part, by PDE9.
- the PDE9-/- non-human mammal or animal cell exhibits the effect to a degree comparable to the PDE9+/+ or PDE9+/- non-human mammal or animal cell, then the effect is mediated by a pathway that does not involve PDE9 signaling.
- an agent is suspected of possibly exerting an effect predominantly via a PDE9 pathway, then the PDE9-/- non-human mammals and animal cells are useful as negative controls to test this hypothesis. If the agent is indeed acting through PDE9, then the PDE9-/- non-human mammals and animal cells, upon administration of the agent, should not demonstrate the same effect observed in the PDE9+/+ non-human mammals or animal cells.
- the genetically modified non-human mammals and animal cells of the invention can also be used to identify genes whose expression is differentially regulated in PDE9+/- or PDE9-/- non-human mammals or animal cells relative to their respective wild type control. Techniques known to those of skill in the art can be used to identify such genes based upon the present description. For example, DNA arrays can be used to identify genes whose expression is differentially regulated in PDE9+/- or PDE9-/- mice to compensate for a deficiency in PDE9 expression. DNA arrays are known to those of skill in the art (see, e.g., Aigner et al., Arthritis and Rheumatism 44: 2777-89, 2001 ; U.S. Pat. No.
- a chemical coupling procedure and an ink jet device may be used to synthesize array elements on the surface of a substrate.
- An array analogous to a dot or slot blot may also be used to arrange and link elements to the surface of a substrate using thermal, UV, chemical, or mechanical bonding procedures.
- a typical array may be produced by hand or using available methods and machines and contain any appropriate number of elements. After hybridization, nonhybridized probes are removed and a scanner used to determine the levels and patterns of fluorescence.
- Full-length cDNAs, expressed sequence tags (ESTs), or fragments thereof may comprise the elements of a microarray. Fragments suitable for hybridization may be selected using software well known in the art such as LASERGENE software (DNASTAR). Full-length cDNAs, ESTs, or fragments thereof corresponding to one of the nucleotide sequences of the present invention, or selected at random from a cDNA library relevant to the present invention, are arranged on an appropriate substrate, e.g., a glass slide.
- the cDNA is fixed to the slide using, e.g., ultra-violet cross-linking followed by thermal and chemical treatments and subsequent drying. Fluorescent probes are prepared and used for hybridization to the elements on the substrate.
- the substrate is analyzed by procedures well known in the art, for example, by scanning and analyzing images of a microarray.
- the genetically modified non-human mammals and animal cells of the invention can also be used to identify proteins whose expression profile or postranslational modification is altered in PDE9+/- or PDE9-/- non-human mammals or animal cells relative to their respective wild type control. Techniques known to those of skill in the art can be used to identify such proteins based upon the present description.
- proteomic assays can be used to identify proteins whose expression profile or postranslational modification is altered in PDE9+/- or PDE9-/- mice to compensate for a deficiency in PDE9 expression.
- Proteomic assays are known to those of skill in the art (see, e.g., Conrads et al., Biochem. Biophys. Res. Commun.
- PDE9 Targeting Vector A targeting vector construct was designed according to the scheme shown in Fig. 1. The construct contained two arms homologous to the murine PDE9 genomic sequence: a 0.9 kb 5' homology arm and a 4.3 kb 3' homology arm. These arms sandwiched a LacZ-Neo construct. DNA containing the targeting construct was inserted into ES R1 cells by electroporation (Deng et al., Dev. Biol. 185: 42-54, 1997; Udy et al., Exp. Cell Res. 231 : 296-301 , 1997). Upon homologous recombination, base pairs 142-175 of the PDE9 cDNA coding sequence shown in Fig.
- ES cells that were neomycin resistant were analyzed by Southern blot to confirm disruption of a PDE9 gene. These targeted ES cells were then used for generation of chimeric mice by injecting the cells into blastocysts and implanting the blastocysts into pseudopregnant female mice (Capecchi et al., Trends Genet. 5: 70, 1989, Hogan et al., Manipulating the Mouse Embryo: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988; and Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E.J.
- mice of each gender remained on D11 mouse chow and the remaining groups of each gender were switched to a diet composed of 58 kcal% fat (D12331 Rodent Diet, Research Diets, Inc., New Brunswick, NJ) for the duration of the 6 week study.
- Body weight was determined on Day 0 and monitored weekly.
- Adipose depot mass was analyzed on Day 0 and at the end of the study, as further described below.
- plasma glucose was determined via retro-orbital blood samples. 25 ⁇ L of blood was added to 100 ⁇ L of 0.025 percent heparinized-saline in microtubes (Denville Scientific, Inc., Metuchen, NJ).
- the tubes were spun at the highest setting in a Beckman Microfuge 12 for 2 minutes. Plasma was collected for plasma glucose and triglyceride determination, as further described below. During the course of the study, body weight and food consumption were assessed, and blood samples were taken at approximately 8 am for plasma glucose and triglyceride measures, as further described below. On the morning of the last day of the study, blood samples were taken via retro-orbital sinus for plasma glucose and triglyceride determination. The mice were then sacrificed and about one milliliter of blood was collected in Microtainer® plasma separator tubes with lithium heparin (Becton-Dickinson, Inc., Franklin Lakes, NJ). The tubes were spun in a Beckman Microfuge 12 at the maximum setting for five minutes.
- Plasma cGMP was measured using the BioTrakTM enzyme- immunoassay system (Amersham, Piscataway, NJ). Plasma insulin was assessed via a similar technique using the Mercodia ELISA Insulin kit supplied by ALPCO (Uppsala, Sweden). All assays were conducted according to each manufacturer's instructions. Quantification of adipose depot mass was done five days prior to the end of the study. To assess the adipose depot mass, 360° radioscopic images of the mice were obtained using a commercially available micro computed tomography (CT) system (MicroCAT ® , ImTek Inc., Oak Ridge, TN) with a high-resolution CCD/phosphor screen detector.
- CT micro computed tomography
- the scanner consisted of a cylindrical diameter/long field view of 36mm/36mm with a spatial resolution of less than 50 ⁇ M.
- the X-ray source was biased at 40 KeV with the anode current set to 0.4 mA.
- Anesthetized mice were placed on a radiotransparent mouse bed in an anatomically correct supine position, caudal end closest to the micro CT with the rostral end held in place against an anesthesia delivery tube.
- An initial radiographic image was acquired at 90° to the plane of the mouse bed to allow correct positioning of the mouse by centering the scan acquisition area at the level of the iliac crest of each mouse. Once correct alignment was assured, each animal was scanned.
- Each scan consisted of 196 individual projections with an exposure time of 250 ⁇ s/projection; total image acquisition time was approximately 12 minutes at 145 ⁇ M resolution.
- Image reconstruction whereby the 196 projections acquired in the micro CT scan of the mouse were manipulated to produce two-dimensional cross sectional images of the mouse, was performed using the MicroCAT ® Reconstruction, Visualization, and Analysis Software (ImTek Inc., Oak Ridge, TN) (Paulus et al., Neoplasia 2: 62-70, 2000). Two sets of reconstructed images per scan were generated for each mouse for the determination of individual fat depot mass. The first set of six reconstructed images provided a montage for the analysis of inguinal and epididymal adipose tissue depot mass.
- the second reconstruction set consisted of nine slices, determined by both intervertebral and midvertebral landmarks, and was used to determine retroperitoneal and mesenteric adipose tissue depot mass.
- reconstructed bitmap images were converted to TIFF images.
- the TIFF images were subsequently analyzed and fat depot mass determined using Scion Image for Windows® (Scion Corporation, Frederick MD). Demarcation lines separating individual fat depots were placed using the paintbrush tool (pixel size #3) and total pixel counts of each adipose region determined by the Scion Image software.
- An upper and lower pixel intensity threshold was chosen, in this study, a look-up-table (LUT) of between 115-187 was determined to be optimal for capturing the adipose depot. Average pixel number between each slice was calculated (slice n +slice n+1 )/2).
- the first factor corrects for specific gravity of glyceryl trioleate, representative of the density of the primary storage form of lipid in adipose tissue, i.e., triglyceride.
- the second factor is the volume per pixel and the third factor converts the resulting mass into mg units.
- mice also demonstrated a 6% decrease in body length.
- the male and female KO mice also demonstrated decreased fat mass in various adipose depots (Fig. 4).
- male KO mice significant decreases were seen in the retroperitoneal and mesenteric adipose depots; in female KO mice, significant decreases were seen in the inguinal, gonadal, and retroperitoneal depots.
- female mice fed a standard chow diet no differences in body weight were observed between KO and wild type mice (Fig.
- mice Female ob/ob mice obtained from Jackson Laboratories (Bar Harbor, ME) were used at 6 to 10 weeks of age. Mice were housed five per cage and allowed free access to water and, initially, to D11 mouse chow. Following a one week acclimation period, mice were switched to a powdered diet (Mouse Breeder/Auto-JL K20 mouse chow, PMI Feeds, Inc., St. Louis, MO) for three days and allowed to adapt to the diet prior to the start of the PDE9 inhibitor dosing period.
- a powdered diet (Mouse Breeder/Auto-JL K20 mouse chow, PMI Feeds, Inc., St. Louis, MO) for three days and allowed to adapt to the diet prior to the start of the PDE9 inhibitor dosing period.
- the PDE9 inhibitor compound (Compound A) was administered in powdered mouse chow that was custom ground (Research Diets, Inc., New Brunswick, NJ) as a compound/chow admixture; compounds were mixed with the chow to achieve consumption of the specified doses ranging from 1-200 mg/kg/day.
- a group consuming darglitazone (1 mg/kg/day) was also included as a positive control.
- Mice were randomly assigned to groups of ten with five mice per cage. Body weight was determined on Day 0 and weekly thereafter. On Day 1 , retro- orbital blood samples were obtained and plasma glucose was determined as previously described.
- Fig. 7 shows a reduced body weight gain in ob/ob mice fed 100 mg/kg/day of the PDE9 inhibitor Compound A as compared to the mice fed either a compound-free control diet or a darglitazone-treated diet.
- Compound A elicited a dose-dependent effect following 2 and 4 days of treatment, both in terms of reducing the normal body weight gain (Fig. 8A) and also in terms of reducing food intake (Fig. 8B).
- the PDE9 effect on food intake could be transient, given that no effect on food intake was observed in the later stages of the study (Fig. 9) with the intermediate dose of 100 mg/kg/day.
- the intermediate dose of 100 mg/kg/day of Compound A also resulted in decreased glucose, triglycerides and fructosamine.
- Representative results are shown in Fig. 10, Fig. 11, and Fig. 12, respectively.
- Both Examples demonstrate that causing a decrease in PDE9 activity is an effective method to reduce body weight and/or body fat, and can be used, e.g., to treat animal patients that are overweight, obese, or suffer from an eating disorder, and can be used in animal food stock species to produce leaner meat.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Chemical & Material Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Veterinary Medicine (AREA)
- Biomedical Technology (AREA)
- Animal Behavior & Ethology (AREA)
- Medicinal Chemistry (AREA)
- Zoology (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Wood Science & Technology (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Biotechnology (AREA)
- General Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Diabetes (AREA)
- Microbiology (AREA)
- Physics & Mathematics (AREA)
- Hematology (AREA)
- Environmental Sciences (AREA)
- Plant Pathology (AREA)
- Biophysics (AREA)
- Developmental Biology & Embryology (AREA)
- Gynecology & Obstetrics (AREA)
- Reproductive Health (AREA)
- Cardiology (AREA)
- Heart & Thoracic Surgery (AREA)
- Obesity (AREA)
- Urology & Nephrology (AREA)
- Animal Husbandry (AREA)
- Emergency Medicine (AREA)
- Vascular Medicine (AREA)
Abstract
L'invention concerne des méthodes de diminution du poids du corps ou de la graisse contenue dans le corps d'un animal, par exemple lors du traitement de patients souffrant de surcharge pondérale ou d'obésité (que ce soient des êtres humains ou des animaux), ou afin de produire de la viande plus maigre chez des animaux d'élevage (par exemple du bétail, des poulets ou des cochons), ainsi que des méthodes de traitement de troubles de l'alimentation (tels que la frénésie alimentaire ou la boulimie) chez des patients qui en ont besoin, par administration d'un inhibiteur de la PDE 9. L'invention concerne également des outils biologiques qui permettent d'approfondir l'étude de la fonction de la PDE 9, c'est-à-dire des souris et des cellules animales génétiquement modifiées ayant une disruption génétique de la PDE 9.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US51621303P | 2003-10-31 | 2003-10-31 | |
PCT/IB2004/003396 WO2005041972A1 (fr) | 2003-10-31 | 2004-10-18 | Inhibition de la phosphodiesterase 9 comme traitement d'etats associes a l'obesite |
Publications (1)
Publication Number | Publication Date |
---|---|
EP1686998A1 true EP1686998A1 (fr) | 2006-08-09 |
Family
ID=34549505
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
EP04769658A Withdrawn EP1686998A1 (fr) | 2003-10-31 | 2004-10-18 | Inhibition de la phosphodiesterase 9 comme traitement d'etats associes a l'obesite |
Country Status (6)
Country | Link |
---|---|
EP (1) | EP1686998A1 (fr) |
JP (1) | JP2007509923A (fr) |
BR (1) | BRPI0416118A (fr) |
CA (1) | CA2543522A1 (fr) |
TW (1) | TW200526783A (fr) |
WO (1) | WO2005041972A1 (fr) |
Families Citing this family (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CA2886885C (fr) | 2011-10-10 | 2019-07-16 | H. Lundbeck A/S | Pde9i ayant un squelette imidazo pyrazinone |
US9434733B2 (en) | 2012-01-26 | 2016-09-06 | H. Lundbeck A/S | PDE9 inhibitors with imidazo triazinone backbone |
SI3865484T1 (sl) | 2015-07-07 | 2024-05-31 | H. Lundbeck A/S | Zaviralec pde9 z imidazo pirazinonsko hrbtenico za zdravljenje perifernih bolezni |
EP4219709A1 (fr) * | 2016-11-03 | 2023-08-02 | Temple University - Of The Commonwealth System of Higher Education | Plasmides d'adn pour la génération rapide de vecteurs de recombinaison homologues pour le développement de lignées cellulaires |
JP2021526134A (ja) | 2018-05-25 | 2021-09-30 | イマラ インク. | 6−[(3S,4S)−4−メチル−l−(ピリミジン−2−イルメチル)ピロリジン−3−イル]−3−テトラヒドロピラン−4−イル−7H−イミダゾ[l,5−A]ピラジン−8−オンの一水和物および結晶形態 |
WO2020076752A1 (fr) * | 2018-10-08 | 2020-04-16 | The Johns Hopkins University | Utilisation d'inhibiteurs de pde9 pour un traitement |
Family Cites Families (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5922595A (en) * | 1997-12-09 | 1999-07-13 | Incyte Pharmaceuticals, Inc. | Cyclic GMP phosphodiesterase |
EP1150674A1 (fr) * | 1999-02-12 | 2001-11-07 | Novo Nordisk A/S | Utilisation de derives de pyrrolidine pour la preparation d'une composition pharmaceutique destinee au traitement ou a la prevention de l'obesite ou a la regulation de l'appetit |
US20020165237A1 (en) * | 2000-08-11 | 2002-11-07 | Fryburg David Albert | Treatment of the insulin resistance syndrome |
US20030114469A1 (en) * | 2001-09-27 | 2003-06-19 | Cohen David Saul | Combinations |
KR20040053210A (ko) * | 2001-11-02 | 2004-06-23 | 화이자 프로덕츠 인크. | Pde9 억제제를 사용한 인슐린 저항 증후군 및 2형당뇨병의 치료 |
US20030181461A1 (en) * | 2002-01-25 | 2003-09-25 | Lautt Wilfred Wayne | Use of phosphodiesterase antagonists to treat insulin resistance |
US20040220186A1 (en) * | 2003-04-30 | 2004-11-04 | Pfizer Inc. | PDE9 inhibitors for treating type 2 diabetes,metabolic syndrome, and cardiovascular disease |
-
2004
- 2004-10-18 CA CA002543522A patent/CA2543522A1/fr not_active Abandoned
- 2004-10-18 EP EP04769658A patent/EP1686998A1/fr not_active Withdrawn
- 2004-10-18 BR BRPI0416118-1A patent/BRPI0416118A/pt not_active IP Right Cessation
- 2004-10-18 WO PCT/IB2004/003396 patent/WO2005041972A1/fr not_active Application Discontinuation
- 2004-10-18 JP JP2006537457A patent/JP2007509923A/ja not_active Withdrawn
- 2004-10-29 TW TW093133068A patent/TW200526783A/zh unknown
Non-Patent Citations (1)
Title |
---|
See references of WO2005041972A1 * |
Also Published As
Publication number | Publication date |
---|---|
CA2543522A1 (fr) | 2005-05-12 |
BRPI0416118A (pt) | 2007-01-02 |
WO2005041972A1 (fr) | 2005-05-12 |
JP2007509923A (ja) | 2007-04-19 |
TW200526783A (en) | 2005-08-16 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Jackson et al. | Defective valvulogenesis in HB‐EGF and TACE‐null mice is associated with aberrant BMP signaling | |
WO2005041972A1 (fr) | Inhibition de la phosphodiesterase 9 comme traitement d'etats associes a l'obesite | |
US20080282364A1 (en) | Phosphodiesterase 9 Inhibition as Treatment for Obesity-Related Conditions | |
EP1980148B1 (fr) | Animal genetiquement modifie et son utilisation | |
CN108300737B (zh) | 一种表型高度一致的恶性淋巴瘤模型的建立方法及其用途 | |
Furuta et al. | Recent innovations in tissue‐specific gene modifications in the mouse | |
JP4851041B2 (ja) | 細胞内のカルシウムイオンの濃度変化をモニターするトランスジェニック非ヒト哺乳動物 | |
US20050144659A1 (en) | Animals and cells containing a mutated alpha2delta gene | |
MXPA06004887A (en) | Phosphodiesterase 9 inhibition as treatment for obesity-related conditions | |
AU741340B2 (en) | Non human transgenic animal in which the expression of the gene coding for insulin is deleted | |
US20030131370A1 (en) | Disruption of the glutathione S-transferase-Omega-1 gene | |
US20030121069A1 (en) | Disruption of the phosphodiesterase 10 gene | |
US20030157076A1 (en) | Disruption of the Akt2 gene | |
EP1321034A1 (fr) | Disruption du gene de phosphodiesterase 10 | |
JPH11155420A (ja) | トランスジェニック動物 | |
US20030106085A1 (en) | Disruption of the prostaglandin E synthase 2 gene | |
JP2003286195A (ja) | Pde11aの活性を調節する剤 | |
JP2006149380A (ja) | 遺伝子改変動物の新規用途 | |
JP2004166596A (ja) | Zaq遺伝子改変動物 | |
JP2003310261A (ja) | Redk遺伝子の破壊 | |
JP2010220615A (ja) | ゲノムが改変されたマウス | |
ZA200404167B (en) | Disruption of the prostaglandin E synthase 2 gene. |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PUAI | Public reference made under article 153(3) epc to a published international application that has entered the european phase |
Free format text: ORIGINAL CODE: 0009012 |
|
17P | Request for examination filed |
Effective date: 20060531 |
|
AK | Designated contracting states |
Kind code of ref document: A1 Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PL PT RO SE SI SK TR |
|
DAX | Request for extension of the european patent (deleted) | ||
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN |
|
18W | Application withdrawn |
Effective date: 20071210 |