WO2009032840A1 - Fluorine- substituted 2 ', 6 ' -dimethyl-l-tyrosine-1, 2,3, 4-tetrahydr0is0quin0line-3-carb0xylic acid peptides (dmt-tic) for use as myu- and delta-opioid receptor probes in - Google Patents
Fluorine- substituted 2 ', 6 ' -dimethyl-l-tyrosine-1, 2,3, 4-tetrahydr0is0quin0line-3-carb0xylic acid peptides (dmt-tic) for use as myu- and delta-opioid receptor probes in Download PDFInfo
- Publication number
- WO2009032840A1 WO2009032840A1 PCT/US2008/075114 US2008075114W WO2009032840A1 WO 2009032840 A1 WO2009032840 A1 WO 2009032840A1 US 2008075114 W US2008075114 W US 2008075114W WO 2009032840 A1 WO2009032840 A1 WO 2009032840A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compound
- salt
- optical isomer
- group
- tissue
- Prior art date
Links
- 101100244562 Pseudomonas aeruginosa (strain ATCC 15692 / DSM 22644 / CIP 104116 / JCM 14847 / LMG 12228 / 1C / PRS 101 / PAO1) oprD gene Proteins 0.000 title claims description 31
- 108700023159 delta Opioid Receptors Proteins 0.000 title claims description 31
- 102000048124 delta Opioid Receptors Human genes 0.000 title claims description 31
- 108090000765 processed proteins & peptides Proteins 0.000 title description 12
- 239000002253 acid Substances 0.000 title description 10
- 108010013218 2',6'-dimethyltyrosyl-1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid Proteins 0.000 title description 5
- 102000004196 processed proteins & peptides Human genes 0.000 title description 3
- 239000000523 sample Substances 0.000 title description 3
- 150000001875 compounds Chemical class 0.000 claims abstract description 143
- 150000003839 salts Chemical class 0.000 claims abstract description 77
- 238000000034 method Methods 0.000 claims abstract description 35
- 210000000056 organ Anatomy 0.000 claims abstract description 30
- 239000003937 drug carrier Substances 0.000 claims abstract description 5
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 4
- -1 optical isomer Chemical class 0.000 claims description 169
- 230000003287 optical effect Effects 0.000 claims description 63
- 210000001519 tissue Anatomy 0.000 claims description 61
- 125000003118 aryl group Chemical group 0.000 claims description 49
- 125000000539 amino acid group Chemical group 0.000 claims description 43
- 239000000203 mixture Substances 0.000 claims description 26
- 102000051367 mu Opioid Receptors Human genes 0.000 claims description 24
- 108020001612 μ-opioid receptors Proteins 0.000 claims description 24
- 125000001153 fluoro group Chemical group F* 0.000 claims description 23
- 125000001072 heteroaryl group Chemical group 0.000 claims description 23
- 125000003588 lysine group Chemical group [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 claims description 21
- 125000005098 aryl alkoxy carbonyl group Chemical group 0.000 claims description 20
- 125000005099 aryl alkyl carbonyl group Chemical group 0.000 claims description 20
- 125000005129 aryl carbonyl group Chemical group 0.000 claims description 19
- 238000002600 positron emission tomography Methods 0.000 claims description 16
- 102000005962 receptors Human genes 0.000 claims description 16
- 108020003175 receptors Proteins 0.000 claims description 16
- 206010028980 Neoplasm Diseases 0.000 claims description 15
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 claims description 15
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 claims description 14
- 125000000613 asparagine group Chemical group N[C@@H](CC(N)=O)C(=O)* 0.000 claims description 12
- 125000000404 glutamine group Chemical group N[C@@H](CCC(N)=O)C(=O)* 0.000 claims description 12
- 125000003282 alkyl amino group Chemical group 0.000 claims description 9
- 125000004663 dialkyl amino group Chemical group 0.000 claims description 9
- JIDDDPVQQUHACU-YFKPBYRVSA-N (2s)-pyrrolidine-2-carbaldehyde Chemical group O=C[C@@H]1CCCN1 JIDDDPVQQUHACU-YFKPBYRVSA-N 0.000 claims description 7
- 239000001257 hydrogen Substances 0.000 claims description 7
- 229910052739 hydrogen Inorganic materials 0.000 claims description 7
- 210000004072 lung Anatomy 0.000 claims description 7
- 201000011510 cancer Diseases 0.000 claims description 6
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 claims description 6
- 125000002470 cystinyl group Chemical group O=C([*])C(N([H])[H])([H])C([H])([H])SSC([H])([H])C([H])(C(O[H])=O)N([H])[H] 0.000 claims description 6
- 125000000291 glutamic acid group Chemical group N[C@@H](CCC(O)=O)C(=O)* 0.000 claims description 6
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 6
- 125000000341 threoninyl group Chemical group [H]OC([H])(C([H])([H])[H])C([H])(N([H])[H])C(*)=O 0.000 claims description 6
- 125000005454 tryptophanyl group Chemical group 0.000 claims description 6
- 125000001493 tyrosinyl group Chemical group [H]OC1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 claims description 6
- 210000000481 breast Anatomy 0.000 claims description 5
- 125000004076 pyridyl group Chemical group 0.000 claims description 5
- 125000000168 pyrrolyl group Chemical group 0.000 claims description 5
- 239000003795 chemical substances by application Substances 0.000 claims description 4
- 210000001072 colon Anatomy 0.000 claims description 4
- 125000003386 piperidinyl group Chemical group 0.000 claims description 4
- 125000000719 pyrrolidinyl group Chemical group 0.000 claims description 4
- 238000004611 spectroscopical analysis Methods 0.000 claims description 3
- 102220497176 Small vasohibin-binding protein_T47D_mutation Human genes 0.000 claims description 2
- 210000003128 head Anatomy 0.000 claims description 2
- 210000001672 ovary Anatomy 0.000 claims description 2
- 210000003079 salivary gland Anatomy 0.000 claims description 2
- 210000002784 stomach Anatomy 0.000 claims description 2
- 238000003325 tomography Methods 0.000 claims description 2
- 125000004191 (C1-C6) alkoxy group Chemical group 0.000 claims 2
- 238000004519 manufacturing process Methods 0.000 claims 2
- 210000005068 bladder tissue Anatomy 0.000 claims 1
- 210000005084 renal tissue Anatomy 0.000 claims 1
- 210000005092 tracheal tissue Anatomy 0.000 claims 1
- 230000015572 biosynthetic process Effects 0.000 description 27
- 238000003786 synthesis reaction Methods 0.000 description 27
- 125000003277 amino group Chemical group 0.000 description 25
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-Dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 21
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 20
- 238000004128 high performance liquid chromatography Methods 0.000 description 18
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 15
- 239000000243 solution Substances 0.000 description 14
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 13
- 238000009472 formulation Methods 0.000 description 13
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 12
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 12
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 12
- 229940024606 amino acid Drugs 0.000 description 11
- 239000002585 base Substances 0.000 description 10
- 125000004432 carbon atom Chemical group C* 0.000 description 10
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 9
- 239000000556 agonist Substances 0.000 description 9
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 9
- 238000005160 1H NMR spectroscopy Methods 0.000 description 8
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 7
- 239000002904 solvent Substances 0.000 description 7
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 6
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 6
- 101100108551 Mus musculus Akr1b7 gene Proteins 0.000 description 6
- 239000004480 active ingredient Substances 0.000 description 6
- 125000000217 alkyl group Chemical group 0.000 description 6
- 238000003556 assay Methods 0.000 description 6
- 229910052757 nitrogen Inorganic materials 0.000 description 6
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 6
- FHZPGIUBXYVUOY-VWGYHWLBSA-N Dermorphin Chemical class C([C@H](N)C(=O)N[C@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CO)C(N)=O)C1=CC=C(O)C=C1 FHZPGIUBXYVUOY-VWGYHWLBSA-N 0.000 description 5
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 5
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 5
- 206010029260 Neuroblastoma Diseases 0.000 description 5
- 230000002378 acidificating effect Effects 0.000 description 5
- 150000001413 amino acids Chemical class 0.000 description 5
- 239000005557 antagonist Substances 0.000 description 5
- 125000003710 aryl alkyl group Chemical group 0.000 description 5
- 230000027455 binding Effects 0.000 description 5
- 230000008499 blood brain barrier function Effects 0.000 description 5
- 210000001218 blood-brain barrier Anatomy 0.000 description 5
- 210000004556 brain Anatomy 0.000 description 5
- 230000000694 effects Effects 0.000 description 5
- 238000002595 magnetic resonance imaging Methods 0.000 description 5
- 239000012074 organic phase Substances 0.000 description 5
- 239000011347 resin Substances 0.000 description 5
- 229920005989 resin Polymers 0.000 description 5
- 241000700198 Cavia Species 0.000 description 4
- 101800002242 Dermorphin Proteins 0.000 description 4
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 4
- KRHYYFGTRYWZRS-UHFFFAOYSA-M Fluoride anion Chemical compound [F-] KRHYYFGTRYWZRS-UHFFFAOYSA-M 0.000 description 4
- KDLHZDBZIXYQEI-UHFFFAOYSA-N Palladium Chemical compound [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 4
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 4
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 4
- 238000004458 analytical method Methods 0.000 description 4
- 238000002591 computed tomography Methods 0.000 description 4
- NPZTUJOABDZTLV-UHFFFAOYSA-N hydroxybenzotriazole Substances O=C1C=CC=C2NNN=C12 NPZTUJOABDZTLV-UHFFFAOYSA-N 0.000 description 4
- 238000003384 imaging method Methods 0.000 description 4
- 238000001727 in vivo Methods 0.000 description 4
- 150000002500 ions Chemical class 0.000 description 4
- 125000004433 nitrogen atom Chemical group N* 0.000 description 4
- 229910052700 potassium Inorganic materials 0.000 description 4
- 239000011591 potassium Substances 0.000 description 4
- 239000000047 product Substances 0.000 description 4
- 239000011541 reaction mixture Substances 0.000 description 4
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 3
- 240000007472 Leucaena leucocephala Species 0.000 description 3
- 239000004472 Lysine Substances 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 239000007832 Na2SO4 Substances 0.000 description 3
- 241000700159 Rattus Species 0.000 description 3
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 3
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 3
- 125000003275 alpha amino acid group Chemical group 0.000 description 3
- 125000003368 amide group Chemical group 0.000 description 3
- 125000002619 bicyclic group Chemical group 0.000 description 3
- 239000012267 brine Substances 0.000 description 3
- 229910052792 caesium Inorganic materials 0.000 description 3
- TVFDJXOCXUVLDH-UHFFFAOYSA-N caesium atom Chemical compound [Cs] TVFDJXOCXUVLDH-UHFFFAOYSA-N 0.000 description 3
- 239000000969 carrier Substances 0.000 description 3
- 210000004027 cell Anatomy 0.000 description 3
- 239000003153 chemical reaction reagent Substances 0.000 description 3
- 239000000460 chlorine Substances 0.000 description 3
- 239000006071 cream Substances 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 239000000839 emulsion Substances 0.000 description 3
- 235000019439 ethyl acetate Nutrition 0.000 description 3
- 238000001914 filtration Methods 0.000 description 3
- 125000005842 heteroatom Chemical group 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 238000001840 matrix-assisted laser desorption--ionisation time-of-flight mass spectrometry Methods 0.000 description 3
- 230000000269 nucleophilic effect Effects 0.000 description 3
- 239000002245 particle Substances 0.000 description 3
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 description 3
- 239000000941 radioactive substance Substances 0.000 description 3
- 229910052938 sodium sulfate Inorganic materials 0.000 description 3
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical compound O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 3
- 239000000725 suspension Substances 0.000 description 3
- 239000003826 tablet Substances 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 238000004809 thin layer chromatography Methods 0.000 description 3
- HPZJMUBDEAMBFI-WTNAPCKOSA-N (D-Ala(2)-mephe(4)-gly-ol(5))enkephalin Chemical compound C([C@H](N)C(=O)N[C@H](C)C(=O)NCC(=O)N(C)[C@@H](CC=1C=CC=CC=1)C(=O)NCCO)C1=CC=C(O)C=C1 HPZJMUBDEAMBFI-WTNAPCKOSA-N 0.000 description 2
- 125000003088 (fluoren-9-ylmethoxy)carbonyl group Chemical group 0.000 description 2
- BBYDXOIZLAWGSL-UHFFFAOYSA-N 4-fluorobenzoic acid Chemical compound OC(=O)C1=CC=C(F)C=C1 BBYDXOIZLAWGSL-UHFFFAOYSA-N 0.000 description 2
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 2
- 108700040991 Ala(2)- deltorphin II Proteins 0.000 description 2
- 206010006187 Breast cancer Diseases 0.000 description 2
- 208000026310 Breast neoplasm Diseases 0.000 description 2
- 241000700199 Cavia porcellus Species 0.000 description 2
- 206010009944 Colon cancer Diseases 0.000 description 2
- 208000001333 Colorectal Neoplasms Diseases 0.000 description 2
- NUNBRHVOPFWRRG-RCEFDBTISA-N Deltorphin B Chemical compound C([C@@H](C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@H](C(=O)N[C@@H](C(C)C)C(=O)NCC(N)=O)C(C)C)NC(=O)[C@@H](C)NC(=O)[C@@H](N)CC=1C=CC(O)=CC=1)C1=CC=CC=C1 NUNBRHVOPFWRRG-RCEFDBTISA-N 0.000 description 2
- 108010010803 Gelatin Proteins 0.000 description 2
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 2
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 2
- 206010027476 Metastases Diseases 0.000 description 2
- 241000699670 Mus sp. Species 0.000 description 2
- SJRJJKPEHAURKC-UHFFFAOYSA-N N-Methylmorpholine Chemical compound CN1CCOCC1 SJRJJKPEHAURKC-UHFFFAOYSA-N 0.000 description 2
- NQRYJNQNLNOLGT-UHFFFAOYSA-N Piperidine Chemical compound C1CCNCC1 NQRYJNQNLNOLGT-UHFFFAOYSA-N 0.000 description 2
- ATUOYWHBWRKTHZ-UHFFFAOYSA-N Propane Chemical compound CCC ATUOYWHBWRKTHZ-UHFFFAOYSA-N 0.000 description 2
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 2
- 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 2
- 229930006000 Sucrose Natural products 0.000 description 2
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 2
- 150000007513 acids Chemical class 0.000 description 2
- 239000013543 active substance Substances 0.000 description 2
- 230000002411 adverse Effects 0.000 description 2
- 239000000443 aerosol Substances 0.000 description 2
- 230000008484 agonism Effects 0.000 description 2
- 229910052784 alkaline earth metal Inorganic materials 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 125000001584 benzyloxycarbonyl group Chemical group C(=O)(OCC1=CC=CC=C1)* 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 229910052799 carbon Inorganic materials 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- 238000005859 coupling reaction Methods 0.000 description 2
- 238000003745 diagnosis Methods 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- ZUOUZKKEUPVFJK-UHFFFAOYSA-N diphenyl Chemical compound C1=CC=CC=C1C1=CC=CC=C1 ZUOUZKKEUPVFJK-UHFFFAOYSA-N 0.000 description 2
- 231100000673 dose–response relationship Toxicity 0.000 description 2
- 239000000796 flavoring agent Substances 0.000 description 2
- 230000006870 function Effects 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 239000008273 gelatin Substances 0.000 description 2
- 229920000159 gelatin Polymers 0.000 description 2
- 235000019322 gelatine Nutrition 0.000 description 2
- 235000011852 gelatine desserts Nutrition 0.000 description 2
- 235000011187 glycerol Nutrition 0.000 description 2
- 239000008187 granular material Substances 0.000 description 2
- 230000007062 hydrolysis Effects 0.000 description 2
- 238000006460 hydrolysis reaction Methods 0.000 description 2
- 210000003405 ileum Anatomy 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 238000000099 in vitro assay Methods 0.000 description 2
- 239000012442 inert solvent Substances 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 230000009401 metastasis Effects 0.000 description 2
- 125000002950 monocyclic group Chemical group 0.000 description 2
- 231100000252 nontoxic Toxicity 0.000 description 2
- 230000003000 nontoxic effect Effects 0.000 description 2
- 150000007524 organic acids Chemical class 0.000 description 2
- 235000005985 organic acids Nutrition 0.000 description 2
- 239000003960 organic solvent Substances 0.000 description 2
- 229910052760 oxygen Inorganic materials 0.000 description 2
- 230000000144 pharmacologic effect Effects 0.000 description 2
- UYWQUFXKFGHYNT-UHFFFAOYSA-N phenylmethyl ester of formic acid Natural products O=COCC1=CC=CC=C1 UYWQUFXKFGHYNT-UHFFFAOYSA-N 0.000 description 2
- NROKBHXJSPEDAR-UHFFFAOYSA-M potassium fluoride Chemical compound [F-].[K+] NROKBHXJSPEDAR-UHFFFAOYSA-M 0.000 description 2
- 230000036515 potency Effects 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 239000003755 preservative agent Substances 0.000 description 2
- 125000003373 pyrazinyl group Chemical group 0.000 description 2
- 125000000714 pyrimidinyl group Chemical group 0.000 description 2
- 230000005855 radiation Effects 0.000 description 2
- 210000000664 rectum Anatomy 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 238000004007 reversed phase HPLC Methods 0.000 description 2
- 229920006395 saturated elastomer Polymers 0.000 description 2
- 230000011664 signaling Effects 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- 229910052708 sodium Inorganic materials 0.000 description 2
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 2
- 239000007921 spray Substances 0.000 description 2
- 125000001424 substituent group Chemical group 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 125000004434 sulfur atom Chemical group 0.000 description 2
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 2
- 125000003039 tetrahydroisoquinolinyl group Chemical group C1(NCCC2=CC=CC=C12)* 0.000 description 2
- 125000006168 tricyclic group Chemical group 0.000 description 2
- 239000003643 water by type Substances 0.000 description 2
- LSSQMISUDUUZCC-DWSYCVKZSA-N (2,5-dioxopyrrolidin-1-yl) 4-fluoranylbenzoate Chemical compound C1=CC([18F])=CC=C1C(=O)ON1C(=O)CCC1=O LSSQMISUDUUZCC-DWSYCVKZSA-N 0.000 description 1
- BTANRVKWQNVYAZ-SCSAIBSYSA-N (2R)-butan-2-ol Chemical compound CC[C@@H](C)O BTANRVKWQNVYAZ-SCSAIBSYSA-N 0.000 description 1
- ZPGDWQNBZYOZTI-SFHVURJKSA-N (2s)-1-(9h-fluoren-9-ylmethoxycarbonyl)pyrrolidine-2-carboxylic acid Chemical compound OC(=O)[C@@H]1CCCN1C(=O)OCC1C2=CC=CC=C2C2=CC=CC=C21 ZPGDWQNBZYOZTI-SFHVURJKSA-N 0.000 description 1
- SWZCTMTWRHEBIN-QFIPXVFZSA-N (2s)-2-(9h-fluoren-9-ylmethoxycarbonylamino)-3-(4-hydroxyphenyl)propanoic acid Chemical compound C([C@@H](C(=O)O)NC(=O)OCC1C2=CC=CC=C2C2=CC=CC=C21)C1=CC=C(O)C=C1 SWZCTMTWRHEBIN-QFIPXVFZSA-N 0.000 description 1
- SJVFAHZPLIXNDH-QFIPXVFZSA-N (2s)-2-(9h-fluoren-9-ylmethoxycarbonylamino)-3-phenylpropanoic acid Chemical compound C([C@@H](C(=O)O)NC(=O)OCC1C2=CC=CC=C2C2=CC=CC=C21)C1=CC=CC=C1 SJVFAHZPLIXNDH-QFIPXVFZSA-N 0.000 description 1
- CNBUSIJNWNXLQQ-NSHDSACASA-N (2s)-3-(4-hydroxyphenyl)-2-[(2-methylpropan-2-yl)oxycarbonylamino]propanoic acid Chemical compound CC(C)(C)OC(=O)N[C@H](C(O)=O)CC1=CC=C(O)C=C1 CNBUSIJNWNXLQQ-NSHDSACASA-N 0.000 description 1
- NUNBRHVOPFWRRG-LJXGPVSSSA-N (4s)-4-[[(2s)-2-[[(2s)-2-[[(2s)-2-amino-3-(4-hydroxyphenyl)propanoyl]amino]propanoyl]amino]-3-phenylpropanoyl]amino]-5-[[(2s)-1-[[(2s)-1-[(2-amino-2-oxoethyl)amino]-3-methyl-1-oxobutan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]amino]-5-oxopentanoic acid Chemical compound C([C@@H](C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@H](C(=O)N[C@@H](C(C)C)C(=O)NCC(N)=O)C(C)C)NC(=O)[C@H](C)NC(=O)[C@@H](N)CC=1C=CC(O)=CC=1)C1=CC=CC=C1 NUNBRHVOPFWRRG-LJXGPVSSSA-N 0.000 description 1
- BWKMGYQJPOAASG-UHFFFAOYSA-N 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid Chemical compound C1=CC=C2CNC(C(=O)O)CC2=C1 BWKMGYQJPOAASG-UHFFFAOYSA-N 0.000 description 1
- DFPYXQYWILNVAU-UHFFFAOYSA-N 1-hydroxybenzotriazole Chemical compound C1=CC=C2N(O)N=NC2=C1.C1=CC=C2N(O)N=NC2=C1 DFPYXQYWILNVAU-UHFFFAOYSA-N 0.000 description 1
- NDKDFTQNXLHCGO-UHFFFAOYSA-N 2-(9h-fluoren-9-ylmethoxycarbonylamino)acetic acid Chemical compound C1=CC=C2C(COC(=O)NCC(=O)O)C3=CC=CC=C3C2=C1 NDKDFTQNXLHCGO-UHFFFAOYSA-N 0.000 description 1
- 108700022183 Ala(2)-MePhe(4)-Gly(5)- Enkephalin Proteins 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
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 241000416162 Astragalus gummifer Species 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- 0 CC=I*(C(CCCC*(C(c(cc1)ccc1N)=O)=C)C(*#C)=O)=C Chemical compound CC=I*(C(CCCC*(C(c(cc1)ccc1N)=O)=C)C(*#C)=O)=C 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 201000009030 Carcinoma Diseases 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- ZAMOUSCENKQFHK-UHFFFAOYSA-N Chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 229920002785 Croscarmellose sodium Polymers 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
- QNAYBMKLOCPYGJ-UWTATZPHSA-N D-alanine Chemical compound C[C@@H](N)C(O)=O QNAYBMKLOCPYGJ-UWTATZPHSA-N 0.000 description 1
- 239000004338 Dichlorodifluoromethane Substances 0.000 description 1
- IAZDPXIOMUYVGZ-WFGJKAKNSA-N Dimethyl sulfoxide Chemical compound [2H]C([2H])([2H])S(=O)C([2H])([2H])[2H] IAZDPXIOMUYVGZ-WFGJKAKNSA-N 0.000 description 1
- 108010092674 Enkephalins Proteins 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- KRHYYFGTRYWZRS-UHFFFAOYSA-N Fluorane Chemical group F KRHYYFGTRYWZRS-UHFFFAOYSA-N 0.000 description 1
- 208000032612 Glial tumor Diseases 0.000 description 1
- 206010018338 Glioma Diseases 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 1
- 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 1
- URLZCHNOLZSCCA-VABKMULXSA-N Leu-enkephalin Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(O)=O)NC(=O)CNC(=O)CNC(=O)[C@@H](N)CC=1C=CC(O)=CC=1)C1=CC=CC=C1 URLZCHNOLZSCCA-VABKMULXSA-N 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 108010093625 Opioid Peptides Proteins 0.000 description 1
- 102000001490 Opioid Peptides Human genes 0.000 description 1
- 102000003840 Opioid Receptors Human genes 0.000 description 1
- 108090000137 Opioid Receptors Proteins 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 229920002873 Polyethylenimine Polymers 0.000 description 1
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 1
- 206010041067 Small cell lung cancer Diseases 0.000 description 1
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 229920001615 Tragacanth Polymers 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 230000001133 acceleration Effects 0.000 description 1
- 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 1
- 239000003513 alkali Substances 0.000 description 1
- 229910052783 alkali metal Inorganic materials 0.000 description 1
- 150000001340 alkali metals Chemical class 0.000 description 1
- 239000003708 ampul Substances 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 150000001450 anions Chemical class 0.000 description 1
- 230000003042 antagnostic effect Effects 0.000 description 1
- 125000002178 anthracenyl group Chemical group C1(=CC=CC2=CC3=CC=CC=C3C=C12)* 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 125000001691 aryl alkyl amino group Chemical group 0.000 description 1
- 125000002102 aryl alkyloxo group Chemical group 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 238000000376 autoradiography Methods 0.000 description 1
- 239000003855 balanced salt solution Substances 0.000 description 1
- 125000003785 benzimidazolyl group Chemical group N1=C(NC2=C1C=CC=C2)* 0.000 description 1
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid group Chemical group C(C1=CC=CC=C1)(=O)O WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 1
- 125000003236 benzoyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C(*)=O 0.000 description 1
- 125000001797 benzyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])* 0.000 description 1
- 239000004305 biphenyl Substances 0.000 description 1
- 235000010290 biphenyl Nutrition 0.000 description 1
- 239000006172 buffering agent Substances 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 125000002837 carbocyclic group Chemical group 0.000 description 1
- 125000002915 carbonyl group Chemical group [*:2]C([*:1])=O 0.000 description 1
- 150000001768 cations Chemical class 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 210000003679 cervix uteri Anatomy 0.000 description 1
- 229910052801 chlorine Inorganic materials 0.000 description 1
- 229940110456 cocoa butter Drugs 0.000 description 1
- 235000019868 cocoa butter Nutrition 0.000 description 1
- 229940075614 colloidal silicon dioxide Drugs 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 201000010989 colorectal carcinoma Diseases 0.000 description 1
- 238000012875 competitive assay Methods 0.000 description 1
- 238000009833 condensation Methods 0.000 description 1
- 230000005494 condensation Effects 0.000 description 1
- 230000008602 contraction Effects 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 239000008120 corn starch Substances 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 229960001681 croscarmellose sodium Drugs 0.000 description 1
- 235000010947 crosslinked sodium carboxy methyl cellulose Nutrition 0.000 description 1
- 238000003350 crude synaptosomal preparation Methods 0.000 description 1
- WZHCOOQXZCIUNC-UHFFFAOYSA-N cyclandelate Chemical compound C1C(C)(C)CC(C)CC1OC(=O)C(O)C1=CC=CC=C1 WZHCOOQXZCIUNC-UHFFFAOYSA-N 0.000 description 1
- 239000000857 delta opiate receptor antagonist Substances 0.000 description 1
- 230000001066 destructive effect Effects 0.000 description 1
- 238000002059 diagnostic imaging Methods 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
- 201000010099 disease Diseases 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- 239000003814 drug Substances 0.000 description 1
- 238000009513 drug distribution Methods 0.000 description 1
- 108010015198 endomorphin 2 Proteins 0.000 description 1
- XIJHWXXXIMEHKW-LJWNLINESA-N endomorphin-2 Chemical compound C([C@H](N)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(N)=O)C1=CC=C(O)C=C1 XIJHWXXXIMEHKW-LJWNLINESA-N 0.000 description 1
- 210000002409 epiglottis Anatomy 0.000 description 1
- 210000003238 esophagus Anatomy 0.000 description 1
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- 230000000763 evoking effect Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 235000019634 flavors Nutrition 0.000 description 1
- ZFKJVJIDPQDDFY-UHFFFAOYSA-N fluorescamine Chemical compound C12=CC=CC=C2C(=O)OC1(C1=O)OC=C1C1=CC=CC=C1 ZFKJVJIDPQDDFY-UHFFFAOYSA-N 0.000 description 1
- 150000004673 fluoride salts Chemical class 0.000 description 1
- 239000006260 foam Substances 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 239000012458 free base Substances 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 235000015203 fruit juice Nutrition 0.000 description 1
- 125000002541 furyl group Chemical group 0.000 description 1
- 238000007429 general method Methods 0.000 description 1
- 239000003365 glass fiber Substances 0.000 description 1
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 1
- 239000010931 gold Substances 0.000 description 1
- 229910052737 gold Inorganic materials 0.000 description 1
- 229910052736 halogen Inorganic materials 0.000 description 1
- 150000002367 halogens Chemical class 0.000 description 1
- 125000004051 hexyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000002883 imidazolyl group Chemical group 0.000 description 1
- 239000012535 impurity Substances 0.000 description 1
- 238000010874 in vitro model Methods 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 229940125425 inverse agonist Drugs 0.000 description 1
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 description 1
- 125000001972 isopentyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 125000002183 isoquinolinyl group Chemical group C1(=NC=CC2=CC=CC=C12)* 0.000 description 1
- 125000001786 isothiazolyl group Chemical group 0.000 description 1
- 125000000842 isoxazolyl group Chemical group 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 210000000867 larynx Anatomy 0.000 description 1
- 239000006193 liquid solution Substances 0.000 description 1
- 239000006210 lotion Substances 0.000 description 1
- 239000007937 lozenge Substances 0.000 description 1
- 201000005296 lung carcinoma Diseases 0.000 description 1
- 159000000003 magnesium salts Chemical class 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 238000002844 melting Methods 0.000 description 1
- 230000008018 melting Effects 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- MYWUZJCMWCOHBA-VIFPVBQESA-N methamphetamine Chemical compound CN[C@@H](C)CC1=CC=CC=C1 MYWUZJCMWCOHBA-VIFPVBQESA-N 0.000 description 1
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 1
- 229940016286 microcrystalline cellulose Drugs 0.000 description 1
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 1
- 239000008108 microcrystalline cellulose Substances 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 210000000214 mouth Anatomy 0.000 description 1
- 239000002324 mouth wash Substances 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 210000003249 myenteric plexus Anatomy 0.000 description 1
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001624 naphthyl group Chemical group 0.000 description 1
- 239000003887 narcotic antagonist Substances 0.000 description 1
- 210000003928 nasal cavity Anatomy 0.000 description 1
- 210000001577 neostriatum Anatomy 0.000 description 1
- 238000002610 neuroimaging Methods 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- FEMOMIGRRWSMCU-UHFFFAOYSA-N ninhydrin Chemical compound C1=CC=C2C(=O)C(O)(O)C(=O)C2=C1 FEMOMIGRRWSMCU-UHFFFAOYSA-N 0.000 description 1
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 1
- 230000009871 nonspecific binding Effects 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
- 125000002347 octyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 239000003402 opiate agonist Substances 0.000 description 1
- 239000003399 opiate peptide Substances 0.000 description 1
- 125000001715 oxadiazolyl group Chemical group 0.000 description 1
- 125000002971 oxazolyl group Chemical group 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 239000004031 partial agonist Substances 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 239000006072 paste Substances 0.000 description 1
- 235000010603 pastilles Nutrition 0.000 description 1
- 125000001147 pentyl group Chemical group C(CCCC)* 0.000 description 1
- 238000010647 peptide synthesis reaction Methods 0.000 description 1
- 230000000737 periodic effect Effects 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 210000003800 pharynx Anatomy 0.000 description 1
- 239000012071 phase Substances 0.000 description 1
- OJMIONKXNSYLSR-UHFFFAOYSA-N phosphorous acid Chemical class OP(O)O OJMIONKXNSYLSR-UHFFFAOYSA-N 0.000 description 1
- 230000000704 physical effect Effects 0.000 description 1
- 230000035479 physiological effects, processes and functions Effects 0.000 description 1
- MXXWOMGUGJBKIW-YPCIICBESA-N piperine Chemical compound C=1C=C2OCOC2=CC=1/C=C/C=C/C(=O)N1CCCCC1 MXXWOMGUGJBKIW-YPCIICBESA-N 0.000 description 1
- 229940075559 piperine Drugs 0.000 description 1
- WVWHRXVVAYXKDE-UHFFFAOYSA-N piperine Natural products O=C(C=CC=Cc1ccc2OCOc2c1)C3CCCCN3 WVWHRXVVAYXKDE-UHFFFAOYSA-N 0.000 description 1
- 235000019100 piperine Nutrition 0.000 description 1
- 239000002798 polar solvent Substances 0.000 description 1
- 125000003367 polycyclic group Chemical group 0.000 description 1
- 239000011698 potassium fluoride Substances 0.000 description 1
- 235000003270 potassium fluoride Nutrition 0.000 description 1
- 229920001592 potato starch Polymers 0.000 description 1
- 239000002244 precipitate Substances 0.000 description 1
- 238000002953 preparative HPLC Methods 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 239000001294 propane Substances 0.000 description 1
- 239000003380 propellant Substances 0.000 description 1
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 125000006239 protecting group Chemical group 0.000 description 1
- 230000002685 pulmonary effect Effects 0.000 description 1
- 125000003226 pyrazolyl group Chemical group 0.000 description 1
- 125000001725 pyrenyl group Chemical group 0.000 description 1
- 125000002098 pyridazinyl group Chemical group 0.000 description 1
- 125000002943 quinolinyl group Chemical group N1=C(C=CC2=CC=CC=C12)* 0.000 description 1
- 230000035484 reaction time Effects 0.000 description 1
- YGSDEFSMJLZEOE-UHFFFAOYSA-M salicylate Chemical compound OC1=CC=CC=C1C([O-])=O YGSDEFSMJLZEOE-UHFFFAOYSA-M 0.000 description 1
- 229960001860 salicylate Drugs 0.000 description 1
- 125000002914 sec-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 239000000741 silica gel Substances 0.000 description 1
- 229910002027 silica gel Inorganic materials 0.000 description 1
- 229910052709 silver Inorganic materials 0.000 description 1
- 239000004332 silver Substances 0.000 description 1
- 208000000587 small cell lung carcinoma Diseases 0.000 description 1
- 210000000813 small intestine Anatomy 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 230000009870 specific binding Effects 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 238000012453 sprague-dawley rat model Methods 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 229910052717 sulfur Inorganic materials 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 210000003568 synaptosome Anatomy 0.000 description 1
- 238000001308 synthesis method Methods 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 235000012222 talc Nutrition 0.000 description 1
- 125000005931 tert-butyloxycarbonyl group Chemical group [H]C([H])([H])C(OC(*)=O)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 150000005621 tetraalkylammonium salts Chemical class 0.000 description 1
- YLQBMQCUIZJEEH-UHFFFAOYSA-N tetrahydrofuran Natural products C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 description 1
- CZDYPVPMEAXLPK-UHFFFAOYSA-N tetramethylsilane Chemical compound C[Si](C)(C)C CZDYPVPMEAXLPK-UHFFFAOYSA-N 0.000 description 1
- 125000003831 tetrazolyl group Chemical group 0.000 description 1
- WROMPOXWARCANT-UHFFFAOYSA-N tfa trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F.OC(=O)C(F)(F)F WROMPOXWARCANT-UHFFFAOYSA-N 0.000 description 1
- 210000001103 thalamus Anatomy 0.000 description 1
- 125000000335 thiazolyl group Chemical group 0.000 description 1
- 239000002562 thickening agent Substances 0.000 description 1
- 125000001544 thienyl group Chemical group 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 238000006257 total synthesis reaction Methods 0.000 description 1
- 210000003437 trachea Anatomy 0.000 description 1
- 239000000196 tragacanth Substances 0.000 description 1
- 235000010487 tragacanth Nutrition 0.000 description 1
- 229940116362 tragacanth Drugs 0.000 description 1
- 125000004306 triazinyl group Chemical group 0.000 description 1
- AQRLNPVMDITEJU-UHFFFAOYSA-N triethylsilane Substances CC[SiH](CC)CC AQRLNPVMDITEJU-UHFFFAOYSA-N 0.000 description 1
- QXTIBZLKQPJVII-UHFFFAOYSA-N triethylsilicon Chemical compound CC[Si](CC)CC QXTIBZLKQPJVII-UHFFFAOYSA-N 0.000 description 1
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 description 1
- 229960004441 tyrosine Drugs 0.000 description 1
- AFVLVVWMAFSXCK-UHFFFAOYSA-N α-cyano-4-hydroxycinnamic acid Chemical compound OC(=O)C(C#N)=CC1=CC=C(O)C=C1 AFVLVVWMAFSXCK-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D217/00—Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
- C07D217/22—Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the nitrogen-containing ring
- C07D217/26—Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
Definitions
- PEPTIDES (DMT-TIC) FOR USE AS MYU- AND DELTA-OPIOID RECEPTOR PROBES IN
- Imaging techniques such as magnetic resonance imaging (MRI), positron emission tomography (PET), and computed tomography (CT scan) provide non-destructive diagnostic examination of tissues or organs.
- PET involves the acquisition of physiologic images based on the detection of positron radiation emitted from a radioactive substance administered to the patient.
- the subsequent images of the human body developed with this technique are used to evaluate a variety of diseases, particularly cancer.
- Targeted delivery of the radioactive substance to the tissue or organ has advantages such as excellent specificity and high binding affinity, and therefore, has received considerable attention from the industry. Accordingly, there is a desire for substances that target and deliver positron- emitting radionuclides to selected organs or tissues.
- the present invention provides fluorine-substituted derivatives of Dmt-Tic (2',6'- dimethyl-L-tyrosine-l,2,3,4-tetrahydroisoquinoline-3-carboxylic acid peptide), for example, a compound of formula (I), optical isomer, or a pharmaceutically acceptable salt thereof:
- the present invention also provides fluorine-substituted derivatives of methoxytyrosine-Tic and dimethoxytyrosine-Tic, for example, a compound of formula (III), optical isomer, or a pharmaceutically acceptable salt thereof:
- the invention further provides a compound of formula (II), optical isomer, or pharmaceutically acceptable salt thereof:
- composition comprising at least one compound of formula (I), (II) or (III) and a pharmaceutically acceptable carrier.
- the Dmt-Tic pharmacophore binds, advantageously, with affinity to ⁇ -opioid and ⁇ -opioid receptors, which are found in certain cancers (e.g., lung, breast, and/or colorectal cancer).
- the fluorine substituent is a radiolabel, 18 F.
- the present invention further provides a method of locating a tumor comprising a ⁇ - or ⁇ -opioid receptor in a subject.
- Figure 1 illustrates a method of synthesis of the compound BG- 137 in an embodiment of the invention.
- Figure 2 illustrates a method of synthesis of the compound BG-139 in an embodiment of the invention.
- the present invention provides agonists and antagonists of ⁇ -opioid and/or ⁇ - opioid receptors that do not pass the blood-brain barrier (BBB) and can be used to locate tumors that comprise ⁇ -opioid and/or ⁇ -opioid receptors.
- BBB blood-brain barrier
- the compounds of the invention are absorbed systemically.
- the present invention is directed to the Dmt-Tic pharmacophore comprising a fluorine-substituent. More specifically, the present invention is directed to a compound of formula (I):
- R 1 is selected from the group consisting of hydrogen, amino, alkylamino, dialkylamino, piperidinyl, pyrrolidinyl, pyrrolyl, and pyridinyl;
- R 2 is one or more amino acid residues; and
- R is selected from the group consisting of aryl, arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, aralkylamido, and heteroaryl, wherein the aryl, heteroaryl, or the "aryl" part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more fluoro or trifluoromethyl groups; an optical isomer thereof; or a pharmaceutically acceptable salt thereof.
- R 1 is selected from the group consisting of amino, alkylamino, and dialkylamino.
- R 2 is one or more amino acid residues, wherein an amino group of the amino acid is bonded to the 3-carboxyl group of the tetrahydroisoquinoline moiety; the amino group that is bonded to the 3-carboxyl group can be an alpha-amino group or another amino such as the side chain amino group of lysine.
- the 3-carboxyl group is bonded to the amino group as an amide group.
- at least one of the amino acid residues has an acidic functionality, such as -COOH or -CONH 2 . When more than one amino acid residue constitutes R , the residues are covalently bonded through amide bonds.
- R 2 comprises 1-6 amino acid residues, preferably 1-3 amino acid residues, and more preferably 1-2 amino acid residues. Any suitable amino acid, natural or synthetic, can be employed. Typically, the one or more amino acid residues are selected from the group consisting of glycinyl, alaninyl, valinyl, leucinyl, isoleucinyl, phenylalaninyl, asparaginyl, glutaminyl, tryptophanyl, prolinyl, serinyl, threoninyl, tyrosinyl, hydroxyprolinyl, cysteinyl, cystinyl, methioninyl, aspartyl, glutamyl, lysinyl, argininyl, and histidinyl.
- R 2 comprises one or more amino acid residues selected from the group consisting of phenylalaninyl, asparaginyl, glutaminyl, serinyl, and lysinyl, and more preferably phenylalaninyl and/or lysinyl.
- the aryl part is substituted with one or more fluoro groups (preferably one) at any suitable ring position.
- the fluoro group is the radiolabel, 18 F.
- R 3 is selected from the group consisting of wherein n is 1 to 6 (e.g., 1, 2, 3, 4, 5, or 6).
- the aryl part is substituted with one or more trifluoromethyl groups (preferably one) at any suitable ring position.
- the aryl part is substituted with one or more trifluoromethyl groups (preferably one) at any suitable ring position.
- the aryl part is substituted with one or more trifluoromethyl groups (preferably one) at any suitable ring position.
- R 'X trifluoromethyl group contains one, two, or three of the radiolabel, F.
- R is selected from the group consisting of
- R 3 is selected from the group consisting of 2-, 3-, or 4- fluorophenylcarbonyl, 2-, 3-, or 4-fiuorobenzylcarbonyl, 2-, 3-, or 4- fluorobenzyloxycarbonyl, 2-, 3-, or 4-fiuorophenylamido, 2-, 3-, or 4-fluorobenzylamido, and 4-, 5-, 6-, or 7-fluoro-l/i-benzimidazole-2-yl ("F-Bid").
- R 3 is selected from the group consisting of 4-fluorophenylcarbonyl, 4-fluorobenzylcarbonyl, 4- fluorobenzyloxycarbonyl, 4-fluorophenylamido, 4-fluorobenzylamido, 5-fluoro-lH- benzimidazole-2-yl ("5F-Bid”), and 6-fluoro-lH-benzimidazole-2-yl ("6F-Bid”).
- R 3 is 4-fluorophenylcarbonyl.
- R is connected to R via any suitable group on R , e.g., a terminal amino or carboxy group on the amino acid residue of R .
- R is connected via the amino group of the amino acid residue, either an alpha-amino group or a side chain amino group.
- R 2 is lysinyl
- R 3 is bonded to the amino group alpha to the carboxy group.
- R could be bonded to the side chain amino group on lysinyl.
- the compound of formula (I) can be chiral or achiral. If the compound is chiral, it can be the R enantiomer, the S enantiomer, or a mixture of both (including a racemic mixture). If more than one chiral center is present, the stereoisomers of the compound of formula (I) can be diastereomers of one another and can include a meso compound.
- the present invention is directed to fluorine-substituted derivatives of methoxytyrosine-Tic and dimethoxytyrosine-Tic (2'-methoxy-L-tyrosine- l,2,3,4-tetrahydroisoquinoline-3-carboxylic acid peptide, 6'-methoxy-L-tyrosine-l,2,3,4- tetrahydroisoquinoline-3-carboxylic acid peptide, and 2',6'-dimethoxy-L-tyrosine-l, 2,3,4- tetrahydroisoquinoline-3-carboxylic acid peptide), for example a compound of formula (III):
- R 1 is selected from the group consisting of hydrogen, amino, alkylamino, dialkylamino, piperidinyl, pyrrolidinyl, pyrrolyl, and pyridinyl;
- R 2 is one or more amino acid residues
- R 3 is selected from the group consisting of aryl, arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, aralkylamido, and heteroaryl, wherein the aryl, heteroaryl, or the "aryl" part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more fluoro or trifluoromethyl groups; an optical isomer thereof; or a pharmaceutically acceptable salt thereof.
- R 1 is selected from the group consisting of amino, alkylamino, and dialkylamino.
- R 2 is one or more amino acid residues, wherein an amino group of the amino acid is bonded to the 3-carboxyl group of the tetrahydroisoquinoline moiety; the amino group that is bonded to the 3-carboxyl group can be an alpha-amino group or another amino such as the side chain amino group of lysine.
- the 3-carboxyl group is bonded to the amino group as an amide group.
- at least one of the amino acid residues has an acidic functionality, such as -COOH or -CONH 2 . When more than one amino acid residue constitutes R 2 , the residues are covalently bonded through amide bonds.
- R 2 comprises 1-6 amino acid residues, preferably 1-3 amino acid residues, and more preferably 1-2 amino acid residues. Any suitable amino acid, natural or synthetic, can be employed. Typically, the one or more amino acid residues are selected from the group consisting of glycinyl, alaninyl, valinyl, leucinyl, isoleucinyl, phenylalaninyl, asparaginyl, glutaminyl, tryptophanyl, prolinyl, serinyl, threoninyl, tyrosinyl, hydroxyprolinyl, cysteinyl, cystinyl, methioninyl, aspartyl, glutamyl, lysinyl, argininyl, and histidinyl.
- R 2 comprises one or more amino acid residues selected from the group consisting of phenylalaninyl, asparaginyl, glutaminyl, serinyl, and lysinyl, and more preferably phenylalaninyl and/or lysinyl.
- the aryl part is substituted with one or more fluoro groups (preferably one) at any suitable ring position.
- the fluoro group is the radiolabel, F.
- R is selected from the group consisting of
- n 1 to 6 (e.g., 1, 2, 3, 4, 5, or 6).
- the aryl part is substituted with one or more trifluoromethyl groups (preferably one) at any suitable ring position.
- the trifluoromethyl group contains one, two, or three of the radiolabel, 18 F.
- R 3 is selected from the group consisting of
- R 3 is selected from the group consisting of 2-, 3-, or 4- fluorophenylcarbonyl, 2-, 3-, or 4-fluorobenzylcarbonyl, 2-, 3-, or 4- fluorobenzyloxycarbonyl, 2-, 3-, or 4-fluorophenylamido, 2-, 3-, or 4-fluorobenzylamido, and 4-, 5-, 6-, or 7-fluoro-l//-benzimidazole-2-yl ("F-Bid").
- R 3 is selected from the group consisting of 4-fluorophenylcarbonyl, 4-fluorobenzylcarbonyl, 4- fluorobenzyloxycarbonyl, 4-fluorophenylarnido, 4-fluorobenzylamido, 5-fluoro-l//- benzimidazole-2-yl ("5F-Bid”), and 6-fluoro-lH-benzimidazole-2-yl ("6F-Bid”).
- R 3 is 4-fluorophenylcarbonyl.
- R 3 is connected to R 2 via any suitable group on R 2 , e.g., a terminal amino or carboxy group on the amino acid residue of R 2 .
- R 3 is connected via the amino group of the amino acid residue, either an alpha-amino group or a side chain amino group.
- R is lysinyl
- R is bonded to the amino group alpha to the carboxy group.
- R 3 could be bonded to the side chain amino group on lysinyl.
- the compound of formula (III) can be chiral or achiral. If the compound is chiral, it can be the R enantiomer, the S enantiomer, or a mixture of both (including a racemic mixture). If more than one chiral center is present, the stereoisomers of the compound of formula (III) can be diastereomers of one another and can include a meso compound. [0028] In another embodiment, the invention is directed to fluorine-substituted derivatives of dermorphin, a ⁇ -selective opioid agonist, of the formula (II):
- R 4 is one or more amino acid residues
- R 5 is selected from the group consisting of aryl, arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, aralkylamido, and heteroaryl, wherein the aryl or heteroaryl group or the "aryl" part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamide, and aralkylamino is substituted with one or more fluoro or trifluoromethyl groups; an optical isomer thereof; or a pharmaceutically acceptable salt thereof.
- R 4 comprises one or more amino acid residues, wherein an amino group of the amino acid is bonded to the carboxyl group of the prolinyl moiety; the amino group that is bonded to the carboxyl group of proline can be an alpha-amino group or another amino group such as the side chain amino group of lysine.
- the carboxyl group is bonded to the amino group as an amide group.
- at least one of the amino acid residues has an acidic functionality, such as -COOH or -CONH 2 . When more than one amino acid residue constitutes R 4 , the residues are covalently bonded through amide bonds.
- R 4 comprises 1-6 amino acid residues, preferably 1-3 amino acid residues, and more preferably 1-2 amino acid residues. Any suitable amino acid, natural or synthetic, can be employed. Typically, the one or more amino acid residues are selected from the group consisting of glycinyl, alaninyl, valinyl, leucinyl, isoleucinyl, phenylalaninyl, asparaginyl, glutaminyl, tryptophanyl, prolinyl, serinyl, threoninyl, tyrosinyl, hydroxyprolinyl, cysteinyl, cystinyl, methioninyl, aspartyl, glutamyl, lysinyl, argininyl, and histidinyl.
- R 4 comprises one or more amino acid residues selected from the group consisting of phenylalaninyl, asparaginyl, glutaminyl, serinyl, and lysinyl, and more preferably phenylalaninyl and/or lysinyl.
- the aryl part is substituted with one or more fluoro groups (preferably one) at any suitable ring position.
- the fluoro group is the radiolabel, 18 F.
- R 5 is selected from the group consisting of
- n 1 to 6 (e.g., 1, 2, 3, 4, 5, or 6).
- the aryl part is substituted with one or more trifluoromethyl groups (preferably one) at any suitable ring position.
- the trifluoromethyl group contains one, two, or three of the radiolabel, F.
- R is selected from the group consisting of
- R is selected from the group consisting of 2-, 3-, or 4- fluorophenylcarbonyl, 2-, 3-, or 4-fluorobenzylcarbonyl, 2-, 3-, or 4- fluorobenzyloxycarbonyl, 2-, 3-, or 4-fluorophenylamido, 2-, 3-, or 4-fluorobenzylamido, and A-, 5-, 6-, or 7-fluoro-l/i-benzimidazole-2-yl ("F-Bid").
- R 5 is selected from the group consisting of 4-fluorophenylcarbonyl, 4-fluorobenzylcarbonyl, 4- fluorobenzyloxycarbonyl, 4-fluorophenylamido, 4-fluorobenzylamido, 5-fluoro-l//- benzimidazole-2-yl ("5F-Bid”), and 6-ftuoro-lH-benzimidazole-2-yl ("6F-Bid”).
- R 5 is 4-fluorophenylcarbonyl.
- R 5 is connected to R 4 via any suitable group on R 4 , e.g., a terminal amino or carboxy group on the amino acid residue of R 4 .
- R 5 is connected via the amino group of the amino acid residue, either alpha-amino group or side chain amino group.
- R 4 is lysinyl, preferably R 5 is bonded to the amino group.
- R 4 could be bonded to the alpha amino group on lysinyl.
- the compound of formula (II) has a molecular weight greater than or equal to about 1,000 g/mol (e.g., 1,000 g/mol or higher, 1,100 g/mol or higher, 1,200 g/mol or higher, 1,500 g/mol or higher).
- compounds of formula (II) can include a charged moiety at, for example, an amino (e.g., - NH 3 + ) or carboxy (e.g., -CO 2 " ) functionality at a functional pH such as 7.4.
- the compound can function as a zwitterion or include a counterion such as a Group I or II cation (e.g., Na + , K + , Mg 2+ , or Ca 2+ ) or a Group VII anion (e.g., Bf, Cl " , or F).
- a counterion such as a Group I or II cation (e.g., Na + , K + , Mg 2+ , or Ca 2+ ) or a Group VII anion (e.g., Bf, Cl " , or F).
- alkyl implies a straight or branched alkyl moiety containing from, for example, 1 to 12 carbon atoms, preferably from 1 to 8 carbon atoms, more preferably from 1 to 6 carbon atoms.
- moieties include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, pentyl, isoamyl, hexyl, octyl, dodecanyl, and the like.
- aryl refers to an unsubstituted or substituted aromatic carbocyclic moiety, as commonly understood in the art, and includes monocyclic and polycyclic aromatics such as, for example, phenyl, biphenyl, naphthyl, anthracenyl, pyrenyl, and the like.
- heteroaryl refers to aromatic 5 or 6 membered monocyclic groups, 9 or 10 membered bicyclic groups, and 11 to 14 membered tricyclic groups which have at least one heteroatom (O, S or N) in at least one of the rings.
- Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom.
- the fused rings completing the bicyclic and tricyclic groups may contain only carbon atoms and may be aromatic, saturated, partially saturated, or unsaturated.
- the nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen atoms may optionally be quaternized.
- Heteroaryl groups which are bicyclic or tricyclic must include at least one fully aromatic ring but the other fused ring or rings may be aromatic or non-aromatic.
- the heteroaryl group may be attached at any available nitrogen or carbon atom of any ring.
- heteroaryl groups are pyridinyl, pyridazinyl, pyrimidyl, pyrazinyl, benzimidazolyl (e.g., lH-benzimidazole-2-yl), triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3,)- and (l,2,4)-triazolyl, pyrazinyl, pyrimidinyl, tetrazolyl, thienyl, isothiazolyl, quinolinyl, isoquinolinyl, thiazolyl, furyl, isoxazolyl, oxadiazolyl, and oxazolyl.
- aralkyl refers to an alkyl group that is substituted with an aryl group.
- the aryl and aralkyl groups are as described herein.
- An example of an aralkyl group is benzyl.
- arylcarbonyl and “arylalkylcarbonyl” refers to the group -C(O)R, in which R is an aryl or aralkyl group, respectively.
- the aryl and aralkyl groups are as described herein.
- An example of an arylcarbonyl group is phenylcarbonyl.
- An example of arylalkylcarbonyl group is benzylcarbonyl.
- aralkyloxycarbonyl refers to an aralkyloxy moiety bound to a carbonyl.
- An example of an aralkyloxycarbonyl is benzyloxycarbonyl.
- alkylamino refers to a group with one hydrogen and one alkyl group directly attached to a trivalent nitrogen atom.
- dialkylamino refers to a group with two of the same or different alkyl groups directly attached to a trivalent nitrogen atom.
- arylamido and aralkylamido refer to the groups -C(O)NHAr and -C(O)NH(CH 2 ) n Ar, respectively, in which Ar is an aryl group as described herein.
- alkyl is as defined herein.
- n is 1 to 6.
- pharmaceutically acceptable salt is meant to include salts of the compound of formula (I) which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.
- base addition salts can be obtained by contacting the free acid form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent.
- pharmaceutically acceptable base addition salts include alkali or alkaline earth metal salts, such as sodium, potassium, calcium, magnesium salts, or ammonium, organic amino, or a similar salt.
- acid addition salts can be obtained by contacting the free base form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
- pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, trifluroacetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, /7-toluenesulfonic, citric, tartaric, methanesulfonic, and the like.
- salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al., "Pharmaceutical Salts," Journal of Pharmaceutical Science, 66: 1-19 (1977)).
- compounds of the present invention can contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
- the neutral forms of the compounds may be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
- the parent form of the compound may differ from the various salt forms in certain physical properties, such as solubility in polar solvents.
- the radiolabel is 18 F.
- 18 F has a half-life (t*0 of 110 minutes, emits ⁇ + particles at an energy of 635 keV, and is 97% abundant.
- F can be obtained from cyclotrons after bombardment of 18 O-enriched water with protons.
- the enriched water containing H- 18 F can be neutralized with a base having a counter-ion that is any alkali metal (M), such as potassium or another monovalent ion, and the water can be evaporated off to give a residue of M- 18 F, which can be taken up in an organic solvent for further use.
- M alkali metal
- the counter- ion is selected to enable the fluoride ion to react rapidly in an organic phase with a halogen. Potassium is generally used as a counter-ion because it is cheaper than cesium. However, with carrier-free F, trivial amounts of counter-ion are required, and the counter-ion cost is minimal.
- Cesium is useful as a counter ion since it is a larger ion with a more diffuse charge. Accordingly, cesium has looser ionic interactions with the small fluoride atom, and therefore does not interfere with the nucleophilic properties of the fluoride ion.
- potassium is preferred to sodium, and, in general, the suitability of a Group Ia metal as a counter-ion in accordance with the present invention increases down the periodic table.
- Group Ib reagents, such as silver also are useful as counter-ions.
- organic phase transfer-type ions such as tetraalkylammonium salts, also can be used as counter-ions.
- Fluoride salts can have a tendency to become hydrated and lose their nucleophilic character.
- the labeling reaction is preferably performed under anhydrous conditions.
- fluoride as potassium fluoride or as a complex with any of the other counter-ions discussed above
- an organic solvent such as acetonitrile or tetrahydrofuran.
- agents which bind to the counter-ion such as Kryptofix 2.2.2 (4,7,13, 16,21,24-hexaoxa-l,10-diazabicyclo[8.8.8]-hexacosane)
- Kryptofix 2.2.2 4,7,13, 16,21,24-hexaoxa-l,10-diazabicyclo[8.8.8]-hexacosane
- the present inventive compounds can be synthesized by any suitable method. See, for example, U.S. Patents 6,916,905 and 6,753,317, Modern Techniques of Peptide and Amino Acid Analysis, John Wiley & Sons, 1981; Bodansky, Principles of Peptide Synthesis, Springer Verlag, 1984).
- compounds of formula (I) or (II) in accordance with embodiments of the invention can be made by starting from a protected amino acid residue (e.g., Lys) and further protecting it. Substituent R 3 is added and then the first protecting group on the amino acid residue can be selectively deprotected.
- R 2 Next additional amino acid residues (R 2 ) are optionally added, then protected Tic is added, and then selectively deprotected. Protected Dmt is added next and then selectively deprotected. The first amino acid residue (R 2 ) can be further deprotected if necessary.
- Compounds of formula (II) can be made from dermorphin. Once R 4 and R 5 have been prepared and coupled, as described above, the remaining peptide sequence can be added using known techniques. For the fluoro substituent on R 3 , the radiolabel 18 F can be used. Specific examples of synthesis methods of the present inventive compounds are set forth in the Examples herein.
- the present invention further provides a pharmaceutical composition
- a pharmaceutical composition comprising at least one compound of formula (I), (II), or (III), optical isomer, or salt thereof and a pharmaceutically acceptable carrier.
- the composition is formulated for human administration.
- Pharmaceutically acceptable carriers are well-known to those of ordinary skill in the art, as are suitable methods of administration. The choice of carrier will be determined, in part, by the particular method used to administer the composition.
- routes of administering a composition are available, and, although more than one route can be used for administration, a particular route can provide a more immediate and more effective reaction than another route. Accordingly, there are a wide variety of suitable formulations of compositions that can be used in the present inventive methods.
- a compound of the present invention can be made into a formulation suitable for parenteral administration, e.g., intravenal, subcutaneous, intraperitoneal, or dural administration.
- a formulation can include aqueous and nonaqueous, isotonic sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and nonaqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
- the formulations can be presented in unit dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water, for injections, immediately prior to use.
- sterile liquid carrier for example, water
- Extemporaneously injectable solutions and suspensions can be prepared from sterile powders, granules, and tablets, as described herein.
- a formulation suitable for oral administration can consist of liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or fruit juice; capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as solid or granules; solutions or suspensions in an aqueous liquid; and oil-in- water emulsions or water-in-oil emulsions.
- diluents such as water, saline, or fruit juice
- capsules, sachets or tablets each containing a predetermined amount of the active ingredient, as solid or granules
- solutions or suspensions in an aqueous liquid and oil-in- water emulsions or water-in-oil emulsions.
- Tablet forms can include one or more of lactose, mannitol, corn starch, potato starch, microcrystalline cellulose, acacia, gelatin, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible carriers.
- a formulation suitable for oral administration can include lozenge forms, which can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier; as well as creams, emulsions, gels, and the like containing, in addition to the active ingredient, such carriers as are known in the art.
- An aerosol formulation suitable for administration via inhalation also can be made.
- the aerosol formulation can be placed into a pressurized acceptable propellant, such as dichlorodifluoromethane, propane, nitrogen, and the like.
- a formulation suitable for topical application can be in the form of creams, ointments, or lotions.
- a formulation for rectal administration can be presented as a suppository with a suitable base comprising, for example, cocoa butter or a salicylate.
- a formulation suitable for vaginal administration can be presented as a pessary, tampon, cream, gel, paste, foam, or spray formula containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
- the dose administered to a subject, particularly a mammal (e.g., human), in the context of the methods of the present invention should be sufficient to produce and image and/or affect a response in the individual over a reasonable time frame.
- the dose will be determined by the potency of the particular compound employed for agonizing/antagonizing the receptor, as well as the body weight and age of the individual.
- the size of the dose also will be determined by the existence of any adverse side effects that may accompany the use of the particular compound employed. It is always desirable, whenever possible, to keep adverse side effects to a minimum.
- the effective amount is used as the preferred endpoint for dosing, the actual dose and schedule can vary, depending on interindividual differences in pharmacokinetics, drug distribution, and metabolism.
- the "effective amount” can be defined, for example, as the blood or tissue level desired in the patient that corresponds to a concentration of one or more compounds according to the invention.
- the "effective amount” for a given compound of the present invention also can vary when the composition of the present invention comprises another active agent or is used in combination with another composition comprising another active agent.
- suitable animal models are available and have been widely implemented for evaluating the in vivo efficacy of such compounds. These models include the hot plate and tail-flick test (see, e.g., U.S. Patent 5,780,589). In vitro models are also available, examples of which are set forth in the Examples herein.
- the dose of the compound of formula (I), (II), or (III), optical isomer or salt thereof desirably comprises about 0.1 mg per kilogram (kg) of the body weight of the mammal (mg/kg) to about 400 mg/kg (e.g., about 0.75 mg/kg, about 5 mg/kg, about 30 mg/kg, about 75 mg/kg, about 100 mg/kg, about 200 mg/kg, or about 300 mg/kg).
- the dose of the compound of formula (I) or (II) comprises about 0.5 mg/kg to about 300 mg/kg (e.g., about 0.75 mg/kg, about 5 mg/kg, about 50 mg/kg, about 100 mg/kg, or about 200 mg/kg), about 10 mg/kg to about 200 mg/kg (e.g., about 25 mg/kg, about 75 mg/kg, or about 150 mg/kg), or about 50 mg/kg to about 100 mg/kg (e.g., about 60 mg/kg, about 70 mg/kg, or about 90 mg/kg).
- antagonist refers to a compound that competes with an endogenous ⁇ -opioid or ⁇ -opioid ligand and inhibits ⁇ - or ⁇ -opioid signaling.
- agonist refers to a compound that competes with the endogenous ⁇ - opioid or ⁇ -opioid ligand and activates or enhances ⁇ - or ⁇ -opioid signaling.
- the present invention further provides a method of locating a ⁇ - and/or ⁇ -opioid receptor that is contained in a tissue or organ of a subject.
- the method comprises the steps of: a) administering a radiolabeled compound of formula (I), (II), or (III), optical isomer or salt thereof to the subject,
- the receptor is a ⁇ -opioid receptor.
- a compound of formula (I) or (III), optical isomer or salt thereof preferably is administered.
- the receptor is a ⁇ -opioid receptor.
- a compound of formula (II), optical isomer or salt thereof preferably is administered.
- the radiolabeled compound of formula (I), (I), or (III), optical isomer or salt thereof is administered in an amount effective to provide an image.
- subject used herein includes animals such as humans, sheep, horses, cattle, pigs, monkeys, dogs, cats, rats, and mice.
- the tissue or organ in the subject to be tested is any tissue or organ that is known to contain a ⁇ - and/or ⁇ -opioid receptor.
- the tissue is peripheral to the brain.
- Such tissues and organs include the entire organ or a tissue sample of a breast, an ovary, a salivary gland, a stomach, a kidney, a colon, a rectum, a cervix, a bladder, a head, a neck, including an esophagus, or the pulmonary system (e.g., a lung, trachea, nasal cavity, mouth, larynx, pharynx, and epiglottis).
- the tissue is from the lung, colon, rectum, or breast. More preferably, the tissue is from the lung.
- the tissue can also include a human cancer cell line known to contain a ⁇ - and/or ⁇ -opioid receptor.
- Such cell lines include NCI-H 146 (small cell lung cancer, ⁇ -opioid receptor), NCI-Hl 87 (non-small cell lung cancer, ⁇ -opioid receptor), SHS Y5 Y (neuroblastoma, ⁇ - and ⁇ -opioid receptors), NS20Y (neuroblastoma, ⁇ -opioid receptor), SK- N-SH (neuroblastoma; ⁇ -opioid receptor), NG108-15 (neuroblastoma-glioma hybrid, ⁇ - opioid receptor), and T47D (breast cancer, ⁇ -opioid receptor).
- Obtaining a diagnostic image of the tissue or organ in step b) typically comprises exposing the tissue or organ in the subject to an energy source, whereupon a diagnostic image of the tissue or organ is obtained.
- the diagnostic image can be, for example, positron emission tomography (PET) image, a magnetic resonance image (MRI), a computerized tomography (CT) scan, single photon emission computed spectroscopy (SPECT) image, or the like.
- PET positron emission tomography
- MRI magnetic resonance image
- CT computerized tomography
- SPECT single photon emission computed spectroscopy
- the diagnostic image can be an MRI.
- the radiolabeled compound of formula (I), (II), or (III), optical isomer or salt thereof distributes in various concentrations to different tissues, and catalyzes the relaxation of protons in the tissues that have been excited by the absorption of radiofrequency energy from a magnetic resonance imager. This acceleration of the rate of relaxation of the excited protons provides for an image of different contrast when the subject is scanned with a magnetic resonance imager.
- the magnetic resonance imager is used to record images at various times, generally either before and after administration of the 18 F labeled compound of formula (I), (II), or (III), or after administration only, and the differences in the images created by the presence of the radiolabeled compound of formula (I) or (II) in tissues are used in diagnosis.
- Guidelines for performing imaging techniques can be found in Stark et al., Magnetic Resonance Imaging, Mosbey Year Book: St. Louis, 1992, hereby incorporated by reference.
- Single Positron Emission Computed Tomography is a non-invasive imaging method to localize the position of a target such as a cancer metastasis, based on radioactive substances that emit gamma radiation when decaying.
- a CT scan provides anatomical detail, such as size and location of the tumor or mass.
- Digital geometry processing is used to generate a three-dimensional image of the internals of an object from a large series of two-dimensional X-ray images taken around a single axis of rotation.
- CT produces a volume of data which can be manipulated, through a process known as windowing, in order to demonstrate various structures based on their ability to block the X-ray beam.
- Combined techniques such as PET/CT and PET/MRI are suitable for use in the invention.
- step b) comprises obtaining a positron emission tomography (PET) image of the tissue or organ.
- PET is a non-invasive imaging method to localize the position of a target such as a cancer metastasis.
- a positron-emitting radionuclide such as 1 F, is introduced, usually by injection, and accumulates in the target tissue or organ. As it decays it emits a positron, which promptly combines with a nearby electron resulting in the simultaneous emission of two identifiable gamma rays in opposite directions.
- a PET scan can provide in vivo physiology such as metabolic detail (e.g., cellular activity) of the tumor or mass. The diagnosis is at a molecular level thereby providing detection of a tumor or mass at an early stage.
- the present invention provides a method of measuring the quantity of a ⁇ - and/or ⁇ -opioid receptor that is contained in a tissue or organ of a subject.
- the method comprises the steps of: a) administering a radiolabeled compound of formula (I), (II), or (III), optical isomer or salt thereof to the subject,
- the receptor is a ⁇ -opioid receptor.
- the receptor is a ⁇ -opioid receptor.
- the radiolabeled compound of formula (I), (II), or (III), optical isomer or salt thereof is administered in an amount effective to provide an image.
- the invention provides a method of locating a tumor comprising a ⁇ -opioid receptor in a mammal in need thereof comprising administering a compound of the formula selected from the group consisting of:
- Crude compounds are purified by preparative reversed-phase HPLC (Waters Delta Prep 4000 system with Waters Prep LC 40 mm Assembly column Cl 8 (30 x 4 cm, 15 ⁇ m particle size)) and eluted at a flow rate of 20 mL/min with mobile phase solvent A (10% acetonitrile + 0.1% TFA in H 2 O, v/v), and a linear gradient from 10 to 60% solvent B (60%, acetonitrile + 0.1% TFA in H 2 O, v/v) in 30 min.
- Analytical HPLC analyses are performed with a Beckman System Gold (Beckman ultrasphere ODS column, 250 x 4.6 mm, 5 ⁇ m particle size).
- Analytical determinations and capacity factor (K') of the products that are used in HPLC in solvents A and B are programmed at flow rate of 1 mL/min with a linear gradient from 0 to 100% B in 25 min. Analogues have less than 1% impurities when monitored at 220 and 254 nm.
- TLC is performed on precoated plates of silica gel F254 (Merck, Darmstadt, Germany): (A) l-butanol/AcOH/H 2 O (3:1:1, v/v/v); (B) CH 2 Cl 2 /toluene/methanol (17:1:2). Ninhydrin (1% ethanol, Merck), fluorescamine (Hoffman-La Roche) and chlorine spray reagents are used. Melting points are determined on a Kofler apparatus and are uncorrected. Optical rotations are assessed at 10 mg/mL in methanol with a Perkin-Elmer 241 polarimeter in a 10 cm water-jacketed cell.
- Boc-Dmt-Tic-£-Lys(4-fluorobenzoyi)-OH is treated with TFA as reported for TFAH-Tic-£--Lys(Z)-OMe: yield 0.09 g (94%); Rf[A) 0.42; HPLC K 4.86; mp 149-151 °C; [Oc] 20 D -18.5; m/z 620 (M+H) + ; 1 H-NMR (DMSO-J 6 ) ⁇ 1.29-1.82 (m, 6H), 2.35 (s, 6H), 2.92- 3.20 (m, 6H), 3.95-4.92 (m, 5H), 6.29 (s, 2H), 6.96-7.93 (m, 8H).
- Boc-Dmt-Tic-Phe-Lys(4-fluorobenzoyl)-OH is treated with TFA as reported for TFAH-Tic- ⁇ -Lys(Z)-OMe: yield 0.08 g (96%); Rf[A) 0.40; HPLC K' 2.87; mp 146-149 °C; [Ot] 20 D +31.1; m/z 161 (M+H) + ; 1 H-NMR (DMSO-Cf 6 ) ⁇ 1.29-1.78 (m, 6H), 2.35 (s, 6H), 2.92- 3.20 (m, 8H), 3.95-4.92 (m, 5H), 6.29 (s, 2H), 6.96-7.93 (m, 13H).
- EXAMPLE 14 This example demonstrates a synthesis of the compound [ 18 F]fluoro-BG-139 in accordance with an embodiment of the invention.
- BG-138 a compound of formula (II), in accordance with an embodiment of the invention.
- Dermorphin is synthesized using Fmoc/t-butyl chemistry with a Syro XP multiple peptide synthesizer (MultiSynTech GmbH, Witten Germany).
- Fmoc-Rink-amide MBHA resin (0.7 mmol/g, 0.150 g) is treated with 40% piperine/DMF and linked with: Fmoc- Lys(Dde)-OH, Fmoc-Pro-OH, Fmoc-Tyr( ⁇ u)-OH, Fmoc-Gly-OH, Fmoc-Phe-OH, Fmoc-D- AIa-OH, Boc-Tyr-OH (4 equiv.; 0.5 M in DMF) by using HOBt (5 equiv.; 0.78 M in DMF), DIC (7 equiv.; 1.09 M in DMF) as the coupling reagent.
- the coupling reaction time is 1.5 h and piperidine (40%)/DMF is used to remove the Fmoc group at each step (20 min).
- the peptide resin is washed with CH 2 Cl 2 and dried in vacuo to yield the protected Dermorphin resin.
- the protected Dermorphin resin is treated with TFA/H 2 O/triethylsilane (9 : 0.5 :
- This example demonstrates a method to determine the equilibrium receptor binding of compounds of formula (I) and (II) in accordance with an embodiment of the invention.
- Opioid receptor affinities are determined under equilibrium conditions (2.5 h at room temperature) in a competition assay using brain P 2 synaptosomal membranes prepared from Sprague-Dawley rats (e.g., Lazarus et al., J. Med. Chem. 1991, 34, 1350-1355; Lazarus et al., Peptides 1993, 14, 21-28). Synaptosomes are preincubated to remove endogenous opioid peptides and stored at -80 °C in buffered 20% glycerol (Lazarus et al., J. Med. Chem. 1991, 34, 1350-1355; Lazarus et al., J Biol. Chem.
- This example demonstrates the functional bioactivity of compounds of the invention in isolated organ preparations in accordance with an embodiment of the invention.
- the isolated tissues are suspended in organ baths containing balanced salt solutions in a physiological buffer, pH 7.5.
- Agonists are tested for the inhibition of electrically evoked contraction and expressed as IC 50 (nM) obtained from the dose-response curves.
- the IC 50 values represent the mean ⁇ SE of five or six separate assays.
- Delta-antagonist potencies in the MVD assay are determined against the ⁇ -agonist deltorphin-II; //-antagonism in the GPI assay uses the //-agonist endomorphin-2, and both are expressed as p ⁇ 2 determined using the Schild Plot (Arunlakshana et al., Br. J. Pharmcol. 1959, 14, 48-58).
- Compounds from Examples 6 and 12 are tested in the electrically stimulated MVD and GPI assays for intrinsic functional bioactivity (Table 2).
- aAgonist activity is expressed as IC 50 obtained from dose-response curves. These values represent the mean ⁇ SE for at least five to six fresh tissue samples.
- pA 2 values of opioid antagonists against the agonists are determined by the method of Kosterlitz and Watt. ⁇ Br J. Pharmacol 1968, 33, 266-276).
- Example 8 The compound of Example 8 is injected into normal mice for brain imaging. The total synthesis time is 120 min and the decay-corrected radiochemical yield of [ F]fluoro- BG-137 is about 30-32% decay starting from [ 18 F]SFB. [ 18 F]Fluoro-BG-137 shows no uptake in mouse brain as it does not cross the blood-brain barrier (BBB). [0106] An in vitro autoradiographic study is also performed by incubating normal rat brain slices with [ 18 F]fluoro-BG-137 without and with the presence of blocking ligands: BG- 137 and UFP-501 [N(Me) 2 -Dmt-Tic-OH].
- the [ F]fluoro-BG-137 shows prominent uptake in the striatum, thalamus, and cortex, which are opioid rich regions. Significant blocking of the uptake by BG- 137 and UFP-501 indicates high specific binding of [ 18 F]fluoro-BG-137 to the ⁇ -opioid receptor.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Peptides Or Proteins (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Disclosed are compounds of formula (I) or pharmaceutically acceptable salts thereof: Formula (I) in which R1, R2, and R3 are described herein. Also disclosed is a pharmaceutical composition comprising at least one compound of formula (I) and a pharmaceutically acceptable carrier. Also disclosed is a method of locating a µ- and/or d-opioid receptor that is contained in a tissue or organ.
Description
FLUORINE-SUBSTITUTED
2 ' , 6 ' -DIMETHYL-L-TYROSINE-1 , 2,3, 4-TETRAHYDR0IS0QUIN0LINE-3-CARB0XYLIC ACID
PEPTIDES (DMT-TIC) FOR USE AS MYU- AND DELTA-OPIOID RECEPTOR PROBES IN
CROSS-REFERENCE TO A RELATED APPLICATION
[0001] This patent application claims the benefit of U.S. Provisional Patent Application No. 60/970,143, filed September 5, 2007, which is incorporated by reference.
BACKGROUND OF THE INVENTION
[0002] Imaging techniques such as magnetic resonance imaging (MRI), positron emission tomography (PET), and computed tomography (CT scan) provide non-destructive diagnostic examination of tissues or organs. In particular, PET involves the acquisition of physiologic images based on the detection of positron radiation emitted from a radioactive substance administered to the patient. The subsequent images of the human body developed with this technique are used to evaluate a variety of diseases, particularly cancer. Targeted delivery of the radioactive substance to the tissue or organ has advantages such as excellent specificity and high binding affinity, and therefore, has received considerable attention from the industry. Accordingly, there is a desire for substances that target and deliver positron- emitting radionuclides to selected organs or tissues.
BRIEF SUMMARY OF THE INVENTION
[0003] The present invention provides fluorine-substituted derivatives of Dmt-Tic (2',6'- dimethyl-L-tyrosine-l,2,3,4-tetrahydroisoquinoline-3-carboxylic acid peptide), for example, a compound of formula (I), optical isomer, or a pharmaceutically acceptable salt thereof:
[0004] The present invention also provides fluorine-substituted derivatives of methoxytyrosine-Tic and dimethoxytyrosine-Tic, for example, a compound of formula (III), optical isomer, or a pharmaceutically acceptable salt thereof:
(III) wherein R1, R2, and R3 are described herein, and wherein R6 is hydrogen and R7 is methoxy or wherein R6 and R7 are both methoxy.
[0005] The invention further provides a compound of formula (II), optical isomer, or pharmaceutically acceptable salt thereof:
H-Tyr-D-Ala-Phe-Gly-Tyr-Pro — R4 — R5
(H) wherein R4 and R5 are described herein.
[0006] Also provided by the present invention is a pharmaceutical composition comprising at least one compound of formula (I), (II) or (III) and a pharmaceutically acceptable carrier.
[0007] The Dmt-Tic pharmacophore binds, advantageously, with affinity to μ-opioid and δ-opioid receptors, which are found in certain cancers (e.g., lung, breast, and/or colorectal cancer). Preferably, the fluorine substituent is a radiolabel, 18F. In an embodiment, since the
1 6 radionuclide F emits a positron, the in vitro and in vivo binding characteristics of the radiolabeled compound of formula (I), (II) or (III) can be assessed using diagnostic imaging,
such as positron emission tomography (PET). Accordingly, the present invention further provides a method of locating a tumor comprising a μ- or δ-opioid receptor in a subject.
BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWING(S)
[0008] Figure 1 illustrates a method of synthesis of the compound BG- 137 in an embodiment of the invention.
[0009] Figure 2 illustrates a method of synthesis of the compound BG-139 in an embodiment of the invention.
DETAILED DESCRIPTION OF THE INVENTION
[0010] The present invention provides agonists and antagonists of μ-opioid and/or δ- opioid receptors that do not pass the blood-brain barrier (BBB) and can be used to locate tumors that comprise μ-opioid and/or δ-opioid receptors. The compounds of the invention are absorbed systemically.
[0011] The present invention is directed to the Dmt-Tic pharmacophore comprising a fluorine-substituent. More specifically, the present invention is directed to a compound of formula (I):
(I) wherein
R1 is selected from the group consisting of hydrogen, amino, alkylamino, dialkylamino, piperidinyl, pyrrolidinyl, pyrrolyl, and pyridinyl; R2 is one or more amino acid residues; and
R is selected from the group consisting of aryl, arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, aralkylamido, and heteroaryl, wherein the aryl, heteroaryl, or the "aryl" part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more fluoro or trifluoromethyl groups; an optical isomer thereof; or a pharmaceutically acceptable salt thereof. [0012] In particular, R1 is selected from the group consisting of amino, alkylamino, and dialkylamino.
[0013] R2 is one or more amino acid residues, wherein an amino group of the amino acid is bonded to the 3-carboxyl group of the tetrahydroisoquinoline moiety; the amino group that is bonded to the 3-carboxyl group can be an alpha-amino group or another amino such as the side chain amino group of lysine. Preferably, the 3-carboxyl group is bonded to the amino group as an amide group. Preferably, at least one of the amino acid residues has an acidic functionality, such as -COOH or -CONH2. When more than one amino acid residue constitutes R , the residues are covalently bonded through amide bonds. For example, R2 comprises 1-6 amino acid residues, preferably 1-3 amino acid residues, and more preferably 1-2 amino acid residues. Any suitable amino acid, natural or synthetic, can be employed. Typically, the one or more amino acid residues are selected from the group consisting of glycinyl, alaninyl, valinyl, leucinyl, isoleucinyl, phenylalaninyl, asparaginyl, glutaminyl, tryptophanyl, prolinyl, serinyl, threoninyl, tyrosinyl, hydroxyprolinyl, cysteinyl, cystinyl, methioninyl, aspartyl, glutamyl, lysinyl, argininyl, and histidinyl. In an embodiment, R2 comprises one or more amino acid residues selected from the group consisting of phenylalaninyl, asparaginyl, glutaminyl, serinyl, and lysinyl, and more preferably phenylalaninyl and/or lysinyl.
[0014] In an embodiment, for R3, the aryl part is substituted with one or more fluoro groups (preferably one) at any suitable ring position. Preferably, the fluoro group is the radiolabel, 18F. For example, R3 is selected from the group consisting of
wherein n is 1 to 6 (e.g., 1, 2, 3, 4, 5, or 6).
[0015] In an embodiment, for R3, the aryl part is substituted with one or more trifluoromethyl groups (preferably one) at any suitable ring position. Preferably, the
I R 'X trifluoromethyl group contains one, two, or three of the radiolabel, F. For example, R is selected from the group consisting of
[0016] In particular embodiments, R3 is selected from the group consisting of 2-, 3-, or 4- fluorophenylcarbonyl, 2-, 3-, or 4-fiuorobenzylcarbonyl, 2-, 3-, or 4- fluorobenzyloxycarbonyl, 2-, 3-, or 4-fiuorophenylamido, 2-, 3-, or 4-fluorobenzylamido, and 4-, 5-, 6-, or 7-fluoro-l/i-benzimidazole-2-yl ("F-Bid"). In certain embodiments, R3 is selected from the group consisting of 4-fluorophenylcarbonyl, 4-fluorobenzylcarbonyl, 4- fluorobenzyloxycarbonyl, 4-fluorophenylamido, 4-fluorobenzylamido, 5-fluoro-lH- benzimidazole-2-yl ("5F-Bid"), and 6-fluoro-lH-benzimidazole-2-yl ("6F-Bid"). Preferably, R3 is 4-fluorophenylcarbonyl.
[0017] Preferably R is connected to R via any suitable group on R , e.g., a terminal amino or carboxy group on the amino acid residue of R . In embodiments, R is connected via the amino group of the amino acid residue, either an alpha-amino group or a side chain amino group. For example, if R2 is lysinyl, preferably R3 is bonded to the amino group alpha to the carboxy group. Alternatively, R could be bonded to the side chain amino group on lysinyl.
[0018] The compound of formula (I) can be chiral or achiral. If the compound is chiral, it can be the R enantiomer, the S enantiomer, or a mixture of both (including a racemic mixture). If more than one chiral center is present, the stereoisomers of the compound of formula (I) can be diastereomers of one another and can include a meso compound.
[0019] Specific compounds of the present invention have the formula
(Ia) (Ib) an optical isomer thereof, or a pharmaceutically acceptable salt (e.g., trifluoroacetic acid) thereof. Preferably, the fluoro is the radiolabel, F. Compounds of formula (I) are expec to be δ-opioid selective, for example, compounds of formula (Ia) and (Ib).
[0020] In other embodiments, the present invention is directed to fluorine-substituted derivatives of methoxytyrosine-Tic and dimethoxytyrosine-Tic (2'-methoxy-L-tyrosine- l,2,3,4-tetrahydroisoquinoline-3-carboxylic acid peptide, 6'-methoxy-L-tyrosine-l,2,3,4- tetrahydroisoquinoline-3-carboxylic acid peptide, and 2',6'-dimethoxy-L-tyrosine-l, 2,3,4- tetrahydroisoquinoline-3-carboxylic acid peptide), for example a compound of formula (III):
(III) wherein
R1 is selected from the group consisting of hydrogen, amino, alkylamino, dialkylamino, piperidinyl, pyrrolidinyl, pyrrolyl, and pyridinyl;
R2 is one or more amino acid residues; and
R3 is selected from the group consisting of aryl, arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, aralkylamido, and heteroaryl, wherein the aryl, heteroaryl, or the "aryl" part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more fluoro or trifluoromethyl groups; an optical isomer thereof; or a pharmaceutically acceptable salt thereof. [0021] In particular, R1 is selected from the group consisting of amino, alkylamino, and dialkylamino.
[0022] R2 is one or more amino acid residues, wherein an amino group of the amino acid is bonded to the 3-carboxyl group of the tetrahydroisoquinoline moiety; the amino group that is bonded to the 3-carboxyl group can be an alpha-amino group or another amino such as the side chain amino group of lysine. Preferably, the 3-carboxyl group is bonded to the amino
group as an amide group. Preferably, at least one of the amino acid residues has an acidic functionality, such as -COOH or -CONH2. When more than one amino acid residue constitutes R2, the residues are covalently bonded through amide bonds. For example, R2 comprises 1-6 amino acid residues, preferably 1-3 amino acid residues, and more preferably 1-2 amino acid residues. Any suitable amino acid, natural or synthetic, can be employed. Typically, the one or more amino acid residues are selected from the group consisting of glycinyl, alaninyl, valinyl, leucinyl, isoleucinyl, phenylalaninyl, asparaginyl, glutaminyl, tryptophanyl, prolinyl, serinyl, threoninyl, tyrosinyl, hydroxyprolinyl, cysteinyl, cystinyl, methioninyl, aspartyl, glutamyl, lysinyl, argininyl, and histidinyl. In an embodiment, R2 comprises one or more amino acid residues selected from the group consisting of phenylalaninyl, asparaginyl, glutaminyl, serinyl, and lysinyl, and more preferably phenylalaninyl and/or lysinyl.
[0023] In an embodiment, for R3, the aryl part is substituted with one or more fluoro groups (preferably one) at any suitable ring position. Preferably, the fluoro group is the radiolabel, F. For example, R is selected from the group consisting of
[0024] In an embodiment, for R3, the aryl part is substituted with one or more trifluoromethyl groups (preferably one) at any suitable ring position. Preferably, the
trifluoromethyl group contains one, two, or three of the radiolabel, 18F. For example, R3 is selected from the group consisting of
[0025] In particular embodiments, R3 is selected from the group consisting of 2-, 3-, or 4- fluorophenylcarbonyl, 2-, 3-, or 4-fluorobenzylcarbonyl, 2-, 3-, or 4- fluorobenzyloxycarbonyl, 2-, 3-, or 4-fluorophenylamido, 2-, 3-, or 4-fluorobenzylamido, and 4-, 5-, 6-, or 7-fluoro-l//-benzimidazole-2-yl ("F-Bid"). In certain embodiments, R3 is selected from the group consisting of 4-fluorophenylcarbonyl, 4-fluorobenzylcarbonyl, 4- fluorobenzyloxycarbonyl, 4-fluorophenylarnido, 4-fluorobenzylamido, 5-fluoro-l//- benzimidazole-2-yl ("5F-Bid"), and 6-fluoro-lH-benzimidazole-2-yl ("6F-Bid"). Preferably, R3 is 4-fluorophenylcarbonyl.
[0026] Preferably R3 is connected to R2 via any suitable group on R2, e.g., a terminal amino or carboxy group on the amino acid residue of R2. In embodiments, R3 is connected via the amino group of the amino acid residue, either an alpha-amino group or a side chain amino group. For example, if R is lysinyl, preferably R is bonded to the amino group alpha to the carboxy group. Alternatively, R3 could be bonded to the side chain amino group on lysinyl.
[0027] The compound of formula (III) can be chiral or achiral. If the compound is chiral, it can be the R enantiomer, the S enantiomer, or a mixture of both (including a racemic
mixture). If more than one chiral center is present, the stereoisomers of the compound of formula (III) can be diastereomers of one another and can include a meso compound. [0028] In another embodiment, the invention is directed to fluorine-substituted derivatives of dermorphin, a μ-selective opioid agonist, of the formula (II):
H-Tyr-D-Ala-Phe-Gly-Tyr-Pro R4 — R5
(H) wherein
R4 is one or more amino acid residues; and
R5 is selected from the group consisting of aryl, arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, aralkylamido, and heteroaryl, wherein the aryl or heteroaryl group or the "aryl" part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamide, and aralkylamino is substituted with one or more fluoro or trifluoromethyl groups; an optical isomer thereof; or a pharmaceutically acceptable salt thereof. [0029] R4 comprises one or more amino acid residues, wherein an amino group of the amino acid is bonded to the carboxyl group of the prolinyl moiety; the amino group that is bonded to the carboxyl group of proline can be an alpha-amino group or another amino group such as the side chain amino group of lysine. Preferably, the carboxyl group is bonded to the amino group as an amide group. Preferably, at least one of the amino acid residues has an acidic functionality, such as -COOH or -CONH2. When more than one amino acid residue constitutes R4, the residues are covalently bonded through amide bonds. For example, R4 comprises 1-6 amino acid residues, preferably 1-3 amino acid residues, and more preferably 1-2 amino acid residues. Any suitable amino acid, natural or synthetic, can be employed. Typically, the one or more amino acid residues are selected from the group consisting of glycinyl, alaninyl, valinyl, leucinyl, isoleucinyl, phenylalaninyl, asparaginyl, glutaminyl, tryptophanyl, prolinyl, serinyl, threoninyl, tyrosinyl, hydroxyprolinyl, cysteinyl, cystinyl, methioninyl, aspartyl, glutamyl, lysinyl, argininyl, and histidinyl. In an embodiment, R4 comprises one or more amino acid residues selected from the group consisting of phenylalaninyl, asparaginyl, glutaminyl, serinyl, and lysinyl, and more preferably phenylalaninyl and/or lysinyl.
[0030] In an embodiment, for R5, the aryl part is substituted with one or more fluoro groups (preferably one) at any suitable ring position. Preferably, the fluoro group is the radiolabel, 18F. For example, R5 is selected from the group consisting of
wherein n is 1 to 6 (e.g., 1, 2, 3, 4, 5, or 6).
[0031] In an embodiment, for R5, the aryl part is substituted with one or more trifluoromethyl groups (preferably one) at any suitable ring position. Preferably, the trifluoromethyl group contains one, two, or three of the radiolabel, F. For example, R is selected from the group consisting of
[0032] In particular embodiments, R is selected from the group consisting of 2-, 3-, or 4- fluorophenylcarbonyl, 2-, 3-, or 4-fluorobenzylcarbonyl, 2-, 3-, or 4- fluorobenzyloxycarbonyl, 2-, 3-, or 4-fluorophenylamido, 2-, 3-, or 4-fluorobenzylamido, and A-, 5-, 6-, or 7-fluoro-l/i-benzimidazole-2-yl ("F-Bid"). In certain embodiments, R5 is selected from the group consisting of 4-fluorophenylcarbonyl, 4-fluorobenzylcarbonyl, 4- fluorobenzyloxycarbonyl, 4-fluorophenylamido, 4-fluorobenzylamido, 5-fluoro-l//- benzimidazole-2-yl ("5F-Bid"), and 6-ftuoro-lH-benzimidazole-2-yl ("6F-Bid"). Preferably, R5 is 4-fluorophenylcarbonyl.
[0033] R5 is connected to R4 via any suitable group on R4, e.g., a terminal amino or carboxy group on the amino acid residue of R4. In embodiments, R5 is connected via the amino group of the amino acid residue, either alpha-amino group or side chain amino group. For example, if R4 is lysinyl, preferably R5 is bonded to the amino group. R4 could be bonded to the alpha amino group on lysinyl.
[0034] So as not to pass the BBB, preferably, the compound of formula (II) has a molecular weight greater than or equal to about 1,000 g/mol (e.g., 1,000 g/mol or higher, 1,100 g/mol or higher, 1,200 g/mol or higher, 1,500 g/mol or higher). Also preferably, compounds of formula (II) can include a charged moiety at, for example, an amino (e.g., - NH3 +) or carboxy (e.g., -CO2 ") functionality at a functional pH such as 7.4. The compound
can function as a zwitterion or include a counterion such as a Group I or II cation (e.g., Na+, K+, Mg2+, or Ca2+) or a Group VII anion (e.g., Bf, Cl", or F). [0035] In a specific embodiment, the compound of formula (II) is
(H) wherein the fluoro is at the 2, 3, or 4 position of the phenyl group. Preferably, the fluoro is at the 4 position of the phenyl group. Compounds of formula (II) are expected to be μ-opioid selective.
[0036] Referring now to terminology used generically herein, the term "alkyl" implies a straight or branched alkyl moiety containing from, for example, 1 to 12 carbon atoms, preferably from 1 to 8 carbon atoms, more preferably from 1 to 6 carbon atoms. Examples of such moieties include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, pentyl, isoamyl, hexyl, octyl, dodecanyl, and the like.
[0037] The term "aryl" refers to an unsubstituted or substituted aromatic carbocyclic moiety, as commonly understood in the art, and includes monocyclic and polycyclic aromatics such as, for example, phenyl, biphenyl, naphthyl, anthracenyl, pyrenyl, and the like. An aryl moiety generally contains from, for example, 6 to 30 carbon atoms, preferably from 6 to 18 carbon atoms, more preferably from 6 to 14 carbon atoms and most preferably from 6 to 10 carbon atoms. It is understood that the term aryl includes carbocyclic moieties that are planar and comprise 4n+2 π electrons, according to Hϋckel's Rule, wherein n = 1, 2, or 3.
[0038] The term "heteroaryl" refers to aromatic 5 or 6 membered monocyclic groups, 9 or 10 membered bicyclic groups, and 11 to 14 membered tricyclic groups which have at least
one heteroatom (O, S or N) in at least one of the rings. Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom. The fused rings completing the bicyclic and tricyclic groups may contain only carbon atoms and may be aromatic, saturated, partially saturated, or unsaturated. The nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen atoms may optionally be quaternized. Heteroaryl groups which are bicyclic or tricyclic must include at least one fully aromatic ring but the other fused ring or rings may be aromatic or non-aromatic. The heteroaryl group may be attached at any available nitrogen or carbon atom of any ring. Illustrative examples of heteroaryl groups are pyridinyl, pyridazinyl, pyrimidyl, pyrazinyl, benzimidazolyl (e.g., lH-benzimidazole-2-yl), triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3,)- and (l,2,4)-triazolyl, pyrazinyl, pyrimidinyl, tetrazolyl, thienyl, isothiazolyl, quinolinyl, isoquinolinyl, thiazolyl, furyl, isoxazolyl, oxadiazolyl, and oxazolyl.
[0039] The term "aralkyl" refers to an alkyl group that is substituted with an aryl group. The aryl and aralkyl groups are as described herein. An example of an aralkyl group is benzyl.
[0040] The terms "arylcarbonyl" and "arylalkylcarbonyl" refers to the group -C(O)R, in which R is an aryl or aralkyl group, respectively. The aryl and aralkyl groups are as described herein. An example of an arylcarbonyl group is phenylcarbonyl. An example of arylalkylcarbonyl group is benzylcarbonyl.
[0041] The term "aralkyloxycarbonyl" refers to an aralkyloxy moiety bound to a carbonyl. An example of an aralkyloxycarbonyl is benzyloxycarbonyl. [0042] The term "alkylamino" refers to a group with one hydrogen and one alkyl group directly attached to a trivalent nitrogen atom. The term "dialkylamino" refers to a group with two of the same or different alkyl groups directly attached to a trivalent nitrogen atom. The terms "arylamido" and "aralkylamido" refer to the groups -C(O)NHAr and -C(O)NH(CH2)nAr, respectively, in which Ar is an aryl group as described herein. The term "alkyl" is as defined herein. The integer n is 1 to 6.
[0043] The term "pharmaceutically acceptable salt" is meant to include salts of the compound of formula (I) which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. When
compounds of formula (I) contain relatively acidic functionalities, base addition salts can be obtained by contacting the free acid form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base addition salts include alkali or alkaline earth metal salts, such as sodium, potassium, calcium, magnesium salts, or ammonium, organic amino, or a similar salt. When compounds of the present invention contain relatively basic functionalities, acid addition salts can be obtained by contacting the free base form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, trifluroacetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, /7-toluenesulfonic, citric, tartaric, methanesulfonic, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al., "Pharmaceutical Salts," Journal of Pharmaceutical Science, 66: 1-19 (1977)). In accordance with embodiments of the invention, compounds of the present invention can contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
[0044] The neutral forms of the compounds may be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner. The parent form of the compound may differ from the various salt forms in certain physical properties, such as solubility in polar solvents.
[0045] Preferably, the radiolabel is 18F. 18F has a half-life (t*0 of 110 minutes, emits β+ particles at an energy of 635 keV, and is 97% abundant. F can be obtained from cyclotrons after bombardment of 18O-enriched water with protons. The enriched water containing H-18F can be neutralized with a base having a counter-ion that is any alkali metal (M), such as potassium or another monovalent ion, and the water can be evaporated off to give a residue of M-18F, which can be taken up in an organic solvent for further use. In general, the counter- ion is selected to enable the fluoride ion to react rapidly in an organic phase with a halogen. Potassium is generally used as a counter-ion because it is cheaper than cesium. However,
with carrier-free F, trivial amounts of counter-ion are required, and the counter-ion cost is minimal.
[0046] Cesium is useful as a counter ion since it is a larger ion with a more diffuse charge. Accordingly, cesium has looser ionic interactions with the small fluoride atom, and therefore does not interfere with the nucleophilic properties of the fluoride ion. For similar reasons, potassium is preferred to sodium, and, in general, the suitability of a Group Ia metal as a counter-ion in accordance with the present invention increases down the periodic table. Group Ib reagents, such as silver, also are useful as counter-ions. Further, organic phase transfer-type ions, such as tetraalkylammonium salts, also can be used as counter-ions. [0047] Fluoride salts can have a tendency to become hydrated and lose their nucleophilic character. To minimize this, the labeling reaction is preferably performed under anhydrous conditions. For example, fluoride (as potassium fluoride or as a complex with any of the other counter-ions discussed above) can be placed in an organic solvent, such as acetonitrile or tetrahydrofuran. With the help of agents which bind to the counter-ion, such as Kryptofix 2.2.2 (4,7,13, 16,21,24-hexaoxa-l,10-diazabicyclo[8.8.8]-hexacosane), the fluoride ion is very nucleophilic in these solvents.
[0048] The present inventive compounds can be synthesized by any suitable method. See, for example, U.S. Patents 6,916,905 and 6,753,317, Modern Techniques of Peptide and Amino Acid Analysis, John Wiley & Sons, 1981; Bodansky, Principles of Peptide Synthesis, Springer Verlag, 1984). In general, compounds of formula (I) or (II) in accordance with embodiments of the invention, can be made by starting from a protected amino acid residue (e.g., Lys) and further protecting it. Substituent R3 is added and then the first protecting group on the amino acid residue can be selectively deprotected. Next additional amino acid residues (R2) are optionally added, then protected Tic is added, and then selectively deprotected. Protected Dmt is added next and then selectively deprotected. The first amino acid residue (R2) can be further deprotected if necessary. Compounds of formula (II) can be made from dermorphin. Once R4 and R5 have been prepared and coupled, as described above, the remaining peptide sequence can be added using known techniques. For the fluoro substituent on R3, the radiolabel 18F can be used. Specific examples of synthesis methods of the present inventive compounds are set forth in the Examples herein. [0049] The present invention further provides a pharmaceutical composition comprising at least one compound of formula (I), (II), or (III), optical isomer, or salt thereof and a
pharmaceutically acceptable carrier. Also, desirably, the composition is formulated for human administration. Pharmaceutically acceptable carriers are well-known to those of ordinary skill in the art, as are suitable methods of administration. The choice of carrier will be determined, in part, by the particular method used to administer the composition. One of ordinary skill in the art will also appreciate that various routes of administering a composition are available, and, although more than one route can be used for administration, a particular route can provide a more immediate and more effective reaction than another route. Accordingly, there are a wide variety of suitable formulations of compositions that can be used in the present inventive methods.
[0050] A compound of the present invention can be made into a formulation suitable for parenteral administration, e.g., intravenal, subcutaneous, intraperitoneal, or dural administration. Such a formulation can include aqueous and nonaqueous, isotonic sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and nonaqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The formulations can be presented in unit dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water, for injections, immediately prior to use. Extemporaneously injectable solutions and suspensions can be prepared from sterile powders, granules, and tablets, as described herein. [0051] A formulation suitable for oral administration can consist of liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or fruit juice; capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as solid or granules; solutions or suspensions in an aqueous liquid; and oil-in- water emulsions or water-in-oil emulsions. Tablet forms can include one or more of lactose, mannitol, corn starch, potato starch, microcrystalline cellulose, acacia, gelatin, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible carriers.
[0052] Similarly, a formulation suitable for oral administration can include lozenge forms, which can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base, such as gelatin and
glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier; as well as creams, emulsions, gels, and the like containing, in addition to the active ingredient, such carriers as are known in the art.
[0053] An aerosol formulation suitable for administration via inhalation also can be made. The aerosol formulation can be placed into a pressurized acceptable propellant, such as dichlorodifluoromethane, propane, nitrogen, and the like.
[0054] A formulation suitable for topical application can be in the form of creams, ointments, or lotions.
[0055] A formulation for rectal administration can be presented as a suppository with a suitable base comprising, for example, cocoa butter or a salicylate. A formulation suitable for vaginal administration can be presented as a pessary, tampon, cream, gel, paste, foam, or spray formula containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
[0056] The dose administered to a subject, particularly a mammal (e.g., human), in the context of the methods of the present invention should be sufficient to produce and image and/or affect a response in the individual over a reasonable time frame. The dose will be determined by the potency of the particular compound employed for agonizing/antagonizing the receptor, as well as the body weight and age of the individual. The size of the dose also will be determined by the existence of any adverse side effects that may accompany the use of the particular compound employed. It is always desirable, whenever possible, to keep adverse side effects to a minimum.
[0057] Since the "effective amount" is used as the preferred endpoint for dosing, the actual dose and schedule can vary, depending on interindividual differences in pharmacokinetics, drug distribution, and metabolism. The "effective amount" can be defined, for example, as the blood or tissue level desired in the patient that corresponds to a concentration of one or more compounds according to the invention. The "effective amount" for a given compound of the present invention also can vary when the composition of the present invention comprises another active agent or is used in combination with another composition comprising another active agent.
[0058] One of ordinary skill in the art can easily determine the appropriate dose, schedule, and method of administration for the exact formulation of the composition being used, in order to achieve the desired "effective amount" in the individual patient. One skilled
in the art also can readily determine and use an appropriate indicator of the "effective amount" of the compound of the present invention by pharmacological end-point analysis. Various general considerations taken into account in determining the "effective amount" are known to those of skill in the art and are described, e.g., in Gilman et al., eds., Goodman And Gilman's: The Pharmacological Bases of Therapeutics, 8th ed., Pergamon Press, 1990; and Remington's Pharmaceutical Sciences, 17th Ed., Mack Publishing Co., Easton, Pa., 1990, each of which is herein incorporated by reference.
[0059] Further, with respect to determining the effective amount in a patient, suitable animal models are available and have been widely implemented for evaluating the in vivo efficacy of such compounds. These models include the hot plate and tail-flick test (see, e.g., U.S. Patent 5,780,589). In vitro models are also available, examples of which are set forth in the Examples herein.
[0060] The dose of the compound of formula (I), (II), or (III), optical isomer or salt thereof desirably comprises about 0.1 mg per kilogram (kg) of the body weight of the mammal (mg/kg) to about 400 mg/kg (e.g., about 0.75 mg/kg, about 5 mg/kg, about 30 mg/kg, about 75 mg/kg, about 100 mg/kg, about 200 mg/kg, or about 300 mg/kg). In another embodiment, the dose of the compound of formula (I) or (II) comprises about 0.5 mg/kg to about 300 mg/kg (e.g., about 0.75 mg/kg, about 5 mg/kg, about 50 mg/kg, about 100 mg/kg, or about 200 mg/kg), about 10 mg/kg to about 200 mg/kg (e.g., about 25 mg/kg, about 75 mg/kg, or about 150 mg/kg), or about 50 mg/kg to about 100 mg/kg (e.g., about 60 mg/kg, about 70 mg/kg, or about 90 mg/kg).
[0061] The term "antagonist," as used herein, refers to a compound that competes with an endogenous δ-opioid or μ-opioid ligand and inhibits δ- or μ-opioid signaling. In contrast, the term "agonist," as used herein, refers to a compound that competes with the endogenous δ- opioid or μ-opioid ligand and activates or enhances δ- or μ-opioid signaling. [0062] Whether an above-described compound functions as an agonist, a partial agonist, an antagonist, a partial antagonist, an inverse agonist, or a mixed agonist/antagonist is set forth, in part, in the Examples herein. Additionally, conventional techniques known to those of ordinary skill in the art can be used to make such determinations. Examples of such techniques include, but are not limited to, the mouse vas deferens in vitro assay of δ-receptors and the guinea pig ileum in vitro assay of μ-receptors as described in the Examples. The
specificity and affinity of the inventive compounds for μ- and δ-opioid receptors can be determined using any suitable method, such as a non-radiolabelled competitive binding assay (see, e.g., Balboni et al., J Med. Chem., 45: 5556-5563 (2002), Lazarus et al., J Med. Chem., 34: 1350-1359 (1991), Salvadori et al., J. Med. Chem., 42: 5010-5019 (1999), and Balboni et al., Bioorg. Med. Chem., 11: 5435-5441 (2003)). Examples of in vivo studies include, but are not limited to, the hot-plate test or tail-flick test, as described in the Example and in the literature (Harris et al., J Pharmacol. Meth, 20: 103-108 (1988); and Sing et al., P. A. Amber (v. 3.0. rev. A), Dept. Pharm. Chem., University of California, San Francisco, 1988). [0063] Certain tumors and carcinomas have been shown to contain high levels of μ- and/or δ-opioid receptors, such as pulmonary carcinomas, including lung carcinoma, colorectal carcinoma, neuroblastomas, and breast cancer (e.g., Madar et al., J Nucl. Med., 48(2): 207-213 (2007); Campa et al., Cancer Research, 56: 1695-1701 (1996); and Rigaudy et al., Cancer Research, 49: 1846-1832 (1989)). Thus, the present invention further provides a method of locating a μ- and/or δ-opioid receptor that is contained in a tissue or organ of a subject. The method comprises the steps of: a) administering a radiolabeled compound of formula (I), (II), or (III), optical isomer or salt thereof to the subject,
b) obtaining a diagnostic image of the tissue or organ,
c) determining the location of radiolabeled compound of formula (I), (II), or (III), optical isomer or salt thereof bound to the tissue or organ; and
d) correlating the location of the bound radiolabeled compound of formula (I), (II), or (III), optical isomer or salt thereof with the location of the μ- and/or δ-opioid receptor in the subject. The method correlates to tumor detection once the μ- and/or δ-opioid receptor has been located in the subject.
[0064] In some embodiments, the receptor is a μ-opioid receptor. In such embodiments, a compound of formula (I) or (III), optical isomer or salt thereof preferably is administered. In other embodiments, the receptor is a δ-opioid receptor. In such embodiments, a compound of formula (II), optical isomer or salt thereof preferably is administered. Preferably, the
radiolabeled compound of formula (I), (I), or (III), optical isomer or salt thereof is administered in an amount effective to provide an image.
[0065] The term "subject" used herein includes animals such as humans, sheep, horses, cattle, pigs, monkeys, dogs, cats, rats, and mice.
[0066] The tissue or organ in the subject to be tested is any tissue or organ that is known to contain a μ- and/or δ-opioid receptor. Preferably the tissue is peripheral to the brain. Such tissues and organs include the entire organ or a tissue sample of a breast, an ovary, a salivary gland, a stomach, a kidney, a colon, a rectum, a cervix, a bladder, a head, a neck, including an esophagus, or the pulmonary system (e.g., a lung, trachea, nasal cavity, mouth, larynx, pharynx, and epiglottis). In preferred embodiments, the tissue is from the lung, colon, rectum, or breast. More preferably, the tissue is from the lung.
[0067] The tissue can also include a human cancer cell line known to contain a μ- and/or δ-opioid receptor. Such cell lines include NCI-H 146 (small cell lung cancer, δ-opioid receptor), NCI-Hl 87 (non-small cell lung cancer, δ-opioid receptor), SHS Y5 Y (neuroblastoma, μ- and δ-opioid receptors), NS20Y (neuroblastoma, δ-opioid receptor), SK- N-SH (neuroblastoma; μ-opioid receptor), NG108-15 (neuroblastoma-glioma hybrid, δ- opioid receptor), and T47D (breast cancer, δ-opioid receptor).
[0068] Obtaining a diagnostic image of the tissue or organ in step b) typically comprises exposing the tissue or organ in the subject to an energy source, whereupon a diagnostic image of the tissue or organ is obtained. The diagnostic image can be, for example, positron emission tomography (PET) image, a magnetic resonance image (MRI), a computerized tomography (CT) scan, single photon emission computed spectroscopy (SPECT) image, or the like.
[0069] The diagnostic image can be an MRI. When administered to a subject, the radiolabeled compound of formula (I), (II), or (III), optical isomer or salt thereof distributes in various concentrations to different tissues, and catalyzes the relaxation of protons in the tissues that have been excited by the absorption of radiofrequency energy from a magnetic resonance imager. This acceleration of the rate of relaxation of the excited protons provides for an image of different contrast when the subject is scanned with a magnetic resonance imager. The magnetic resonance imager is used to record images at various times, generally either before and after administration of the 18F labeled compound of formula (I), (II), or (III),
or after administration only, and the differences in the images created by the presence of the radiolabeled compound of formula (I) or (II) in tissues are used in diagnosis. Guidelines for performing imaging techniques can be found in Stark et al., Magnetic Resonance Imaging, Mosbey Year Book: St. Louis, 1992, hereby incorporated by reference. [0070] Single Positron Emission Computed Tomography (SPECT) is a non-invasive imaging method to localize the position of a target such as a cancer metastasis, based on radioactive substances that emit gamma radiation when decaying.
[0071] A CT scan provides anatomical detail, such as size and location of the tumor or mass. Digital geometry processing is used to generate a three-dimensional image of the internals of an object from a large series of two-dimensional X-ray images taken around a single axis of rotation. CT produces a volume of data which can be manipulated, through a process known as windowing, in order to demonstrate various structures based on their ability to block the X-ray beam. Combined techniques such as PET/CT and PET/MRI are suitable for use in the invention.
[0072] Preferably, step b) comprises obtaining a positron emission tomography (PET) image of the tissue or organ. PET is a non-invasive imaging method to localize the position of a target such as a cancer metastasis. In PET, 511 keV gamma photons produced during positron annihilation decay are detected. A positron-emitting radionuclide, such as 1 F, is introduced, usually by injection, and accumulates in the target tissue or organ. As it decays it emits a positron, which promptly combines with a nearby electron resulting in the simultaneous emission of two identifiable gamma rays in opposite directions. These are detected by a PET camera and give very precise indication of their origin. A PET scan can provide in vivo physiology such as metabolic detail (e.g., cellular activity) of the tumor or mass. The diagnosis is at a molecular level thereby providing detection of a tumor or mass at an early stage.
[0073] Since PET is a quantitative tool, the present invention provides a method of measuring the quantity of a μ- and/or δ-opioid receptor that is contained in a tissue or organ of a subject. The method comprises the steps of: a) administering a radiolabeled compound of formula (I), (II), or (III), optical isomer or salt thereof to the subject,
b) obtaining a positron emission tomography (PET) image of the tissue or organ.
c) determining the amount of radiolabeled compound of formula (I), (II), or (III), optical isomer or salt thereof bound to the tissue or organ; and
d) correlating the amount of the bound radiolabeled compound of formula (I), (II), or (III), optical isomer or salt thereof with the quantity of μ- and/or δ-opioid receptor in the subject. In some embodiments, the receptor is a μ-opioid receptor. In other embodiments, the receptor is a δ-opioid receptor. Preferably, the radiolabeled compound of formula (I), (II), or (III), optical isomer or salt thereof is administered in an amount effective to provide an image.
[0074] In a specific embodiment, the invention provides a method of locating a tumor comprising a δ-opioid receptor in a mammal in need thereof comprising administering a compound of the formula selected from the group consisting of:
(Ia) (Ib) an optical isomer thereof, or a pharmaceutically acceptable salt (e.g., trifluoroacetic acid) thereof.
[0075] The following examples further illustrate the invention but, of course, should not be construed as in any way limiting its scope.
ABBREVIATIONS
Bid lif-benzimidazole-2-yl
Boc tert-butyloxycarbonyl
DAMGO [D-Ala2,Λ/-Me-Phe4,Gly-ol5]enkephalin
DEL C deltorphin II (H-Tyr-D-Ala-Phe-Asp-Val- VaI-GIy-NH2)
DMF iV,jV-dimethylformamide
DMSOd6 hexadeuteriodimethyl sulfoxide
Dmt 2',6'-dimethyl-L-tyrosine
GPI guinea-pig ileum
HOBt 1-hydroxybenzotriazole
HPLC high performance liquid chromatography
MALDI-TOF matrix assisted laser desorption ionization time-of-flight
MPE maximum possible effect
MVD mouse vas deferens
NMM 4-methylmorpholine pA2 negative log of the molar concentration required to double the agonist concentration to achieve the original response
TFA trifluoroacetic acid
Tic 1 ,2,3 ,4-tetrahydroisoquinoline-3 -carboxylic acid
TIP(P) H-Tyr-Tic-Phe-(Phe)-OH
TLC thin-layer chromatography
WSC 1 -ethyl-3-[3'-dimethyl)aminopropyl]-carbodiimide hydrochloride
Z benzyloxycarbonyl
[0076] Crude compounds are purified by preparative reversed-phase HPLC (Waters Delta Prep 4000 system with Waters Prep LC 40 mm Assembly column Cl 8 (30 x 4 cm, 15 μm particle size)) and eluted at a flow rate of 20 mL/min with mobile phase solvent A (10% acetonitrile + 0.1% TFA in H2O, v/v), and a linear gradient from 10 to 60% solvent B (60%, acetonitrile + 0.1% TFA in H2O, v/v) in 30 min. Analytical HPLC analyses are performed with a Beckman System Gold (Beckman ultrasphere ODS column, 250 x 4.6 mm, 5 μm particle size). Analytical determinations and capacity factor (K') of the products that are used in HPLC in solvents A and B are programmed at flow rate of 1 mL/min with a linear gradient from 0 to 100% B in 25 min. Analogues have less than 1% impurities when monitored at 220 and 254 nm.
[0077] TLC is performed on precoated plates of silica gel F254 (Merck, Darmstadt, Germany): (A) l-butanol/AcOH/H2O (3:1:1, v/v/v); (B) CH2Cl2/toluene/methanol (17:1:2).
Ninhydrin (1% ethanol, Merck), fluorescamine (Hoffman-La Roche) and chlorine spray reagents are used. Melting points are determined on a Kofler apparatus and are uncorrected. Optical rotations are assessed at 10 mg/mL in methanol with a Perkin-Elmer 241 polarimeter in a 10 cm water-jacketed cell. Molecular weights of the compounds are determined by a MALDI-TOF analysis (Hewlett Packard G2025 A LD-TOF system mass spectrometer) and α-cyano-4-hydroxycinnamic acid as a matrix. 1H NMR (δ) spectra are measured, when not specified, in DMSO-cfc solution using a Bruker AC-200 spectrometer, and peak positions are given in parts per million downfield from tetramethylsilane as internal standard. [0078] All experiments with animals are carried out according to protocols approved by and on file with the NIEHS Animal Care and Use Committee (ACUC).
EXAMPLE 1
[0079] This example demonstrates a synthesis of TFAH-Tic-ε-Lys(Z)-OMe useful in the synthesis of a compound in accordance with an embodiment of the invention. See Figure 1. [0080] Boc-Tic-£-Lys(Z)-OMe (Balboni et al., Bioorg. Med. Chem. 2007, 15, 3143-3151) (1.67 g, 3.02 mmol) is treated with TFA (2 mL) for 0.5 h at room temperature. Et2O/Pe (1:1, v/v) are added to the solution until the product precipitates out of solution: yield 1.58 g (92%); Rf(A) 0.49; HPLC K' 4.96; mp 118-120 0C; [α]20 D -19.6; m/z 454 (M+H)+.
EXAMPLE 2
[0081] This example demonstrates a synthesis of Boc-Dmt-Tic-ε-Ly S(Z)-OMe useful in the synthesis of a compound in accordance with an embodiment of the invention. See Figure 1.
[0082] To a solution of Boc-Dmt-OH (0.22 g, 0.71 mmol) and TFA H-Tic-e-Lys(Z)-OMe (0.4 g, 0.71 mmol) in DMF (10 mL) at 0 °C, NMM (0.08 mL, 0.71 mmol), HOBt (0.12 g, 0.78 mmol), and WSC (0.15 g, 0.78 mmol) are added. The reaction mixture is stirred for 3 h at 0 0C and 24 h at room temperature. After DMF is evaporated, the residue is dissolved in EtOAc and washed with citric acid (10% in H2O), NaHCO3 (5% in H2O), and brine. The organic phase is dried (Na2SO4) and evaporated to dryness. The residue is precipitated from Et2O/Pe (1:9, v/v): yield 0.46 g (87%); Rf[B) 0.91; HPLC K 7.30; mp 133-135 0C; [α]20 D -
17.5; m/z 746 (M+H)+; 1H-NMR (DMSO-(I6) δ 1.29-1.90 (m, 15H), 2.35 (s, 6H), 2.92-3.20 (m, 6H), 3.67 (s, 3H), 4.41-5.34 (m, 7H), 6.29 (s, 2H), 6.96-7.19 (m, 9H).
EXAMPLE 3
[0083] This example demonstrates a synthesis of Boc-Dmt-Tic-s-Lys-OMe useful in the synthesis of a compound in accordance with an embodiment of the invention. See Figure 1. [0084] To a solution of Boc-Dmt-Tic-£-Lys(Z)-OMe (0.37 g, 0.5 mmol) in methanol (30 mL) is added Pd/C (10%, 0.1 g). H2 is bubbled for 1 h at room temperature. After filtration, the solution is evaporated to dryness. The residue is precipitated from Et2OZPe (1 :9, v/v): yield 0.27 g (90%); Rf[B) 0.74; HPLC K 5.06; mp 139-141 °C; [α]20 D -18.1; m/z 612 (M+H)+.
EXAMPLE 4
[0085] This example demonstrates a synthesis of Boc-Dmt-Tic-£i-Lys(4- fluorobenzoyl)-OMe useful in the synthesis of a compound in accordance with an embodiment of the invention. See Figure 1.
[0086] To a solution of Boc-Dmt-Tic-£-Lys-OMe (0.43 g, 0.7 mmol) and 4-fluorobenzoic acid (0.1 g, 0.7 mmol) in DMF (10 mL) at 0 °C, HOBt (0.12 g, 0.77 mmol), and WSC (0.15 g, 0.77 mmol) are added. The reaction mixture is stirred for 3 h at 0 0C and 24 h at room temperature. After DMF is evaporated, the residue is dissolved in EtOAc and washed with citric acid (10% in H2O), NaHCO3 (5% in H2O), and brine. The organic phase is dried (Na2SO4) and evaporated to dryness. The residue is precipitated from Et2OZPe (1 :9, v/v): yield 0.45 g (88%); Rf[B) 0.79; HPLC K 5.25; mp 134-136 °C; [α]20 D -17.3; m/z 734 (M+H)+; 1H-NMR (DMSO-^) δ 1.29-1.94 (m, 15H), 2.35 (s, 6H), 2.92-3.20 (m, 6H), 3.67 (s, 3H), 4.41-4.92 (m, 5H), 6.29 (s, 2H), 6.96-7.93 (m, 8H).
EXAMPLE 5
[0087] This example demonstrates a synthesis of Boc-Dmt-Tic-£-Lys(4-fluorobenzoyi)- OH useful in the synthesis of a compound in accordance with an embodiment of the invention. See Figure 1.
[0088] To a solution of Boc-Dmt-Tic-£-Lys(4-fluorobenzoyl)-OMe (0.51 g, 0.7 mmol) in ethanol (10 niL) at room temperature, IN NaOH (1.1 mL, 1.1 mmol) is added. The reaction mixture is stirred for 4 h at room temperature. After ethanol is evaporated, the residue is dissolved in EtOAc and washed with citric acid (10% in H2O) and brine. The organic phase is dried (Na2SO4) and evaporated to dryness. The residue is precipitated from Et2O/Pe (1 :9, v/v): yield 0.45 g (90%); RJ[B) 0.75; HPLC K 5.14; mp 141-143 °C; [α]20 D -17.9; m/z 720 (M+H)+.
EXAMPLE 6
[0089] This example demonstrates a synthesis of TFAH-Dmt-Tic-£-Lys(4- fluorobenzoyl)-OH (BG-137) in accordance with an embodiment of the invention. See Figure 1.
[0090] Boc-Dmt-Tic-£-Lys(4-fluorobenzoyi)-OH is treated with TFA as reported for TFAH-Tic-£--Lys(Z)-OMe: yield 0.09 g (94%); Rf[A) 0.42; HPLC K 4.86; mp 149-151 °C; [Oc]20 D -18.5; m/z 620 (M+H)+; 1H-NMR (DMSO-J6) δ 1.29-1.82 (m, 6H), 2.35 (s, 6H), 2.92- 3.20 (m, 6H), 3.95-4.92 (m, 5H), 6.29 (s, 2H), 6.96-7.93 (m, 8H).
EXAMPLE 7
[0091] This example demonstrates a synthesis of Boc-Dmt-Tic-f-Lys-OH useful in the synthesis of a compound in accordance with an embodiment of the invention. See Figure 1. [0092] To a solution of Boc-Dmt-Tic-ε-Lys-OMe (0.09 g, 0.15 mmol; Example 3) in ethanol (10 mL) at room temperature, IN NaOH (0.23 mL, 0.23 mmol) is added. The reaction mixture is stirred for 4 h at room temperature. After ethanol is evaporated, the residue is dissolved in solvent B and directly purified by preparative HPLC as reported above in general methods: yield 0.1 g (92%); Rf[B) 0.67; HPLC K 4.73; mp 146-148 0C; [α]20 D - 18.4; m/z 598 (M+H)+; 1H-NMR (DMSO-^) δ 1.29-1.78 (m, 15H), 2.35 (s, 6H), 2.92-3.49 (m, 7H), 4.41-4.92 (m, 4H), 6.29 (s, 2H), 6.96-7.02 (m, 4H).
EXAMPLE 8 [0093] This example demonstrates a synthesis of the compound [ F]fluoro-BG-137
in accordance with an embodiment of the invention.
[0094] Boc-Dmt-Tic-ε-Lys-OH (Example 7) is coupled with N-succinimidyl-4- [l FJfluorobenzoate under slightly basic conditions at 60 0C for 40 min. The resulting product is deprotected with TFA, and then purified by HPLC to yield [18F]fluoro-BG-137. Yield: 3.2 mg.
EXAMPLE 9
[0095] This example demonstrates a synthesis of Boc-Dmt-Tic-Phe-Lys-OMe useful in the synthesis of a compound in accordance with an embodiment of the invention. See Figure
2.
[0096] To a solution of Boc-Dmt-Tic-Phe-Lys(Z)-OMe (0.71 g, 0.8 mmol) in methanol
(30 mL) Pd/C (10%, 0.2 g) is added, and H2 is bubbled for 1 h at room temperature. After filtration, the solution is evaporated to dryness. The residue is precipitated from Et2O/Pe
(1:9, v/v): yield 0.58 g (88%); Rf[B) 0.81; HPLC K 5.24; mp 144-146 °C; [α]20 D +35.2; m/z
759 (M+H)4
EXAMPLE 10
[0097] This example demonstrates a synthesis of Boc-Dmt-Tic-Phe-Lys(4- fluorobenzoyl)-OMe useful in the synthesis of a compound in accordance with an embodiment of the invention. See Figure 2.
[0098] This compound is obtained by condensation of Boc-Dmt-Tic-Phe-Lys-OMe with 4-fluorobenzoic acid via WSC/HOBt as reported for Boc-Dmt-Tic-ε-Lys(4-fluorobenzoyl)- OMe: yield 0.23 g (85%); RflB) 0.78; HPLC K' 5.10; mp 135-137 0C; [α]20 D +30.6; m/z 881 (M+H)+; 1H-NMR (OMSO-d6) δ 1.29-1.90 (m, 15H), 2.35 (s, 6H), 2.92-3.20 (m, 8H), 3.67 (s, 3H), 4.41-4..92 (m, 6H), 6.29 (s, 2H), 6.96-7.93 (m, 13H).
EXAMPLE I l
[0099] This example demonstrates a synthesis of Boc-Dmt-Tic-Phe-Lys(4- fluorobenzoyl)-OH useful in the synthesis of a compound in accordance with an embodiment of the invention. See Figure 2.
[00100] This compound is obtained by hydrolysis of Boc-Dmt-Tic-Phe-Lys(4- fluorobenzoyl)-OMe with IN NaOH as reported for Boc-Dmt-Tic-£-Lys(4-fluorobenzoyl)-
OH: yield 0.15 g (91%); Rf[B) 0.75; HPLC K 5.0; mp 139-141 °C; [α]20 D +31.4; m/z 867
(M+H)+.
EXAMPLE 12
[00101] This example demonstrates a synthesis of TF A H-Dmt-Tic-Phe-Lys(4- fluorobenzoyl)-OH (BG-139) in accordance with an embodiment of the invention. See Figure 2.
[00102] Boc-Dmt-Tic-Phe-Lys(4-fluorobenzoyl)-OH is treated with TFA as reported for TFAH-Tic-^-Lys(Z)-OMe: yield 0.08 g (96%); Rf[A) 0.40; HPLC K' 2.87; mp 146-149 °C; [Ot]20 D +31.1; m/z 161 (M+H)+; 1H-NMR (DMSO-Cf6) δ 1.29-1.78 (m, 6H), 2.35 (s, 6H), 2.92- 3.20 (m, 8H), 3.95-4.92 (m, 5H), 6.29 (s, 2H), 6.96-7.93 (m, 13H).
EXAMPLE 13
[00103] This example demonstrates a synthesis of Boc-Dmt-Tic-Phe-Lys-OH useful in the synthesis of a compound in accordance with an embodiment of the invention. See Figure 2. [00104] The title compound is obtained by hydrolysis of Boc-Dmt-Tic-Phe-Lys-OMe (Example 9) with IN NaOH as reported for Boc-Dmt-Tic-^-Lys-OH: yield 0.06 g (86%); Rf[B) 0.65; HPLC K! 4.75; mp 140-142 °C; [α]20 D +36.1; m/z 745 (M+H)+; 1H-NMR (DMSO-^) δ 1.29-1.78 (m, 15H), 2.35 (s, 6H), 2.92-3.17 (m, 8H), 4.41-4.92 (m, 6H), 6.29 (s, 2H), 6.96-7.21 (m, 9H).
EXAMPLE 14 [00105] This example demonstrates a synthesis of the compound [18F]fluoro-BG-139
in accordance with an embodiment of the invention.
[00106] Boc-Dmt-Tic-Phe-Lys-OH (Example 13) is coupled with N-succinimidyl-4- [18F]fluorobenzoate under slightly basic conditions at 60 0C for 40 min. The resulting product is deprotected with TFA, and then purified by HPLC to yield [18F]ftuoro-BG-139.
EXAMPLE 15
[00107] This example demonstrates a synthesis of BG-138
BG-138 a compound of formula (II), in accordance with an embodiment of the invention. [00108] Dermorphin is synthesized using Fmoc/t-butyl chemistry with a Syro XP multiple peptide synthesizer (MultiSynTech GmbH, Witten Germany). Fmoc-Rink-amide MBHA resin (0.7 mmol/g, 0.150 g) is treated with 40% piperine/DMF and linked with: Fmoc- Lys(Dde)-OH, Fmoc-Pro-OH, Fmoc-Tyr(Φu)-OH, Fmoc-Gly-OH, Fmoc-Phe-OH, Fmoc-D- AIa-OH, Boc-Tyr-OH (4 equiv.; 0.5 M in DMF) by using HOBt (5 equiv.; 0.78 M in DMF), DIC (7 equiv.; 1.09 M in DMF) as the coupling reagent. The coupling reaction time is 1.5 h and piperidine (40%)/DMF is used to remove the Fmoc group at each step (20 min). The
peptide resin is washed with CH2Cl2 and dried in vacuo to yield the protected Dermorphin resin.
[00109] The protected Dermorphin resin is treated with TFA/H2O/triethylsilane (9 : 0.5 :
0.5 v/v) (10 ml/0.150 g of resin) for 1.5 h at room temperature. After filtration of the exhausted resin, the solvent is concentrated in vacuo, and the residue is triturated with diethyl ether. The crude linear peptide is purified by preparative reverse-phase HPLC to yield a powder after lyophilization.
EXAMPLE 16
[00110] This example demonstrates a method to determine the equilibrium receptor binding of compounds of formula (I) and (II) in accordance with an embodiment of the invention.
[0100] Opioid receptor affinities are determined under equilibrium conditions (2.5 h at room temperature) in a competition assay using brain P2 synaptosomal membranes prepared from Sprague-Dawley rats (e.g., Lazarus et al., J. Med. Chem. 1991, 34, 1350-1355; Lazarus et al., Peptides 1993, 14, 21-28). Synaptosomes are preincubated to remove endogenous opioid peptides and stored at -80 °C in buffered 20% glycerol (Lazarus et al., J. Med. Chem. 1991, 34, 1350-1355; Lazarus et al., J Biol. Chem. 1989, 264, 3047-3050). Each compound is analyzed in duplicate assays using five to eight dosages and three to five independent repetitions with different synaptosomal preparations (n values are listed in Table 1 in parenthesis and results are mean ± SE). Unlabeled compound (2 μM) is used to determine non-specific binding in the presence of 1.9 nM [3H]deltorphin II (45.0 Ci/mmol, Perkin Elmer, Boston, MA; KΌ = 1.4 nM) for ^-opioid receptors and 3.5 nM [3H]DAMGO (50.0 Ci/mmol), Amersham Bioscience, Buckinghamshire, U. K.; K^ = 1.5 nM) for //-opioid receptors. Glass fiber filters (Whatman GFC) are soaked in 0.1% polyethylenimine in order to enhance the signal-to-noise ratio of the bound radiolabeled-synaptosome complex, and the filters are washed thrice in ice-cold buffered BSA (Lazarus et al., J Med. Chem. 1991, 34, 1350-1355). The affinity constants (K\) are calculated according to Cheng and Prusoff (Biochem. Pharmacol. 1973, 22, 3099-3108). Receptor binding and functional bioactivities are reported in Table 1.
Table 1. Receptor binding affinities.
Receptor affinity" Selectivity
a: the K{ values (nM) are determined according to Chang et al., Biochem. Pharmacol. 1973, 22, 3099-3108
EXAMPLE 17
[0101] This example demonstrates the functional bioactivity of compounds of the invention in isolated organ preparations in accordance with an embodiment of the invention. [0102] The myenteric plexus longitudinal muscle preparations (2-3 cm segments) from the small intestine of male Hartley strain guinea pigs (GPI) measure μ-opioid receptor agonism, and a single mouse vas deferens (MVD) is used to determine <S-opioid receptor agonism as described previously (Sasaki et al., Bioorg. Med. Chem. 2003, 11, 675-678). The isolated tissues are suspended in organ baths containing balanced salt solutions in a physiological buffer, pH 7.5. Agonists are tested for the inhibition of electrically evoked contraction and expressed as IC50 (nM) obtained from the dose-response curves. The IC50 values represent the mean ± SE of five or six separate assays. Delta-antagonist potencies in
the MVD assay are determined against the ^-agonist deltorphin-II; //-antagonism in the GPI assay uses the //-agonist endomorphin-2, and both are expressed as pΛ2 determined using the Schild Plot (Arunlakshana et al., Br. J. Pharmcol. 1959, 14, 48-58). [0103] Compounds from Examples 6 and 12 are tested in the electrically stimulated MVD and GPI assays for intrinsic functional bioactivity (Table 2).
Table 2. Functional bioactivities.
aAgonist activity is expressed as IC50 obtained from dose-response curves. These values represent the mean ± SE for at least five to six fresh tissue samples. bThe pA2 values of opioid antagonists against the agonists are determined by the method of Kosterlitz and Watt. {Br J. Pharmacol 1968, 33, 266-276).
EXAMPLE 18
[0104] This example demonstrates rat brain autoradiography of a compound of formula (I) in accordance with an embodiment of the invention.
[0105] The compound of Example 8 is injected into normal mice for brain imaging. The total synthesis time is 120 min and the decay-corrected radiochemical yield of [ F]fluoro- BG-137 is about 30-32% decay starting from [18F]SFB. [18F]Fluoro-BG-137 shows no uptake in mouse brain as it does not cross the blood-brain barrier (BBB). [0106] An in vitro autoradiographic study is also performed by incubating normal rat brain slices with [18F]fluoro-BG-137 without and with the presence of blocking ligands: BG- 137 and UFP-501 [N(Me)2-Dmt-Tic-OH].
[0107] The [ F]fluoro-BG-137 shows prominent uptake in the striatum, thalamus, and cortex, which are opioid rich regions. Significant blocking of the uptake by BG- 137 and UFP-501 indicates high specific binding of [18F]fluoro-BG-137 to the δ-opioid receptor.
[0108] All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
[0109] The use of the terms "a" and "an" and "the" and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms "comprising," "having," "including," and "containing" are to be construed as open-ended terms (i.e., meaning "including, but not limited to,") unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
[0110] Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible
variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.
Claims
1. A compound of formula (I) :
(I) wherein
R1 is selected from the group consisting of hydrogen, amino, alkylamino, dialkylamino, piperidinyl, pyrrolidinyl, pyrrolyl, and pyridinyl;
R2 is one or more amino acid residues; and
R3 is selected from the group consisting of aryl, arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, aralkylamido, and heteroaryl; wherein the aryl, heteroaryl, or the aryl part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more fluoro or trifiuoromethyl groups; an optical isomer thereof; or a pharmaceutically acceptable salt thereof.
2. The compound, optical isomer, or salt of claim 1, wherein R1 is selected from the group consisting of amino, alkylamino, and dialkylamino.
3. The compound, optical isomer, or salt of claim 1 or 2, wherein R is one or more amino acid residues selected from the group consisting of glycinyl, alaninyl, valinyl, leucinyl, isoleucinyl, phenylalaninyl, asparaginyl, glutaminyl, tryptophanyl, prolinyl, serinyl, threoninyl, tyrosinyl, hydroxyprolinyl, cysteinyl, cystinyl, methioninyl, aspartyl, glutamyl, lysinyl, argininyl, and histidinyl.
4. The compound, optical isomer, or salt of any of claims 1-3, wherein R2 is one or more amino acid residues selected from the group consisting of phenylalaninyl, asparaginyl, glutaminyl, serinyl, and lysinyl.
5. The compound, optical isomer, or salt of any of claims 1-4, wherein the aryl, heteroaryl, or the aryl part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more fluoro groups.
6. The compound, optical isomer, or salt of claim 5, wherein R3 is selected from the group consisting of 2-, 3-, or 4-fluorophenylcarbonyl, 2-, 3-, or 4-fluorobenzylcarbonyl, 2-, 3-, or 4-fluorobenzyloxycarbonyl, 2-, 3-, or 4-fluorophenylamido, 2-, 3-, or A- fluorobenzylamido, and 4-, 5-, 6-, or 7-fluoro-l H-benzimidazole-2-yl.
7. The compound, optical isomer, or salt of claim 5, wherein R3 is selected from the group consisting of 4-fluorophenylcarbonyl, 4-fluorobenzylcarbonyl, A- fluorobenzyloxycarbonyl, 4-fluorophenylamido, 4-fluorobenzylamido, 5-fluoro-lH- benzimidazole-2-yl, and 6-fiuoro-lH-benzimidazole-2-yl.
8. The compound, optical isomer, or salt of any of claims 1-4, wherein the aryl, heteroaryl, or the aryl part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more trifluoromethyl groups.
9. The compound, optical isomer, or salt of claim 8, wherein R is selected from the group consisting of 2-, 3-, or 4-trifluoromethylphenylcarbonyl, 2-, 3-, or A- trifluoromethylbenzylcarbonyl, 2-, 3-, or 4-trifluoromethylbenzyloxycarbonyl, 2-, 3-, or A- trifluoromethylphenylamido, 2-, 3-, or 4-trifluoromethylbenzylamido, and A-, 5-, 6-, or 7- trifluoromethyl- 1 H-benzimidazole-2-yl.
10. The compound, optical isomer, or salt of claim 8, wherein R is selected from the group consisting of 4-trifluoromethylphenylcarbonyl, 4-trifluoromethylbenzylcarbonyl, A- trifluoromethylbenzyloxycarbonyl, 4-trifluoromethylphenylamido, A- trifluoromethylbenzylamido, 5 -trifluoromethyl- 1 H-benzimidazole-2-yl, and 6- trifiuoromethyl- 1 H-benzimidazole-2-yl.
11. The compound, optical isomer, or salt of any of claims 1 -7, that is selected from the group consisting of
12. A compound of formula (III) :
(III) wherein
R1 is selected from the group consisting of hydrogen, amino, alkylamino, dialkylamino, piperidinyl, pyrrolidinyl, pyrrolyl, and pyridinyl;
R is one or more amino acid residues; and
R3 is selected from the group consisting of aryl, arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, aralkylamido, and heteroaryl, wherein the aryl, heteroaryl, or the aryl part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more fluoro or trifluoromethyl groups;
R6 is hydrogen and R7 is C1-C6 alkoxy or R6 and R7 are both C1-C6 alkoxy; an optical isomer thereof; or a pharmaceutically acceptable salt thereof.
13. The compound, optical isomer, or salt of claim 12, wherein R1 is selected from the group consisting of amino, alkylamino, and dialkylamino.
14. The compound, optical isomer, or salt of claim 12 or 13, wherein R is one or more amino acid residues selected from the group consisting of glycinyl, alaninyl, valinyl, leucinyl, isoleucinyl, phenylalaninyl, asparaginyl, glutaminyl, tryptophanyl, prolinyl, serinyl, threoninyl, tyrosinyl, hydroxyprolinyl, cysteinyl, cystinyl, methioninyl, aspartyl, glutamyl, lysinyl, argininyl, and histidinyl.
15. The compound, optical isomer, or salt of any of claims 12-14, wherein R is one or more amino acid residues selected from the group consisting of phenylalaninyl, asparaginyl, glutaminyl, serinyl, and lysinyl.
16. The compound, optical isomer, or salt of any of claims 12-15, wherein the aryl, heteroaryl, or the aryl part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more fluoro groups.
17. The compound, optical isomer, or salt of claim 16, wherein R3 is selected from the group consisting of 2-, 3-, or 4-fluorophenylcarbonyl, 2-, 3-, or 4-fluorobenzylcarbonyl, 2-, 3-, or 4-fluorobenzyloxycarbonyl, 2-, 3-, or 4-fluorophenylamido, 2-, 3-, or A- fluorobenzylamido, and A-, 5-, 6-, or 7-fluoro-lH-benzimidazole-2-yl.
18. The compound, optical isomer, or salt of claim 16, wherein R is selected from the group consisting of 4-fluorophenylcarbonyl, 4-fluorobenzylcarbonyl, A- fluorobenzyloxycarbonyl, 4-fluorophenylamido, 4-fiuorobenzylamido, 5-fluoro-lH- benzimidazole-2-yl, and 6-fluoro-lH-benzimidazole-2-yl.
19. The compound, optical isomer, or salt of any of claims 1-4, wherein the aryl, heteroaryl, or the aryl part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more trifluoromethyl groups
20. The compound, optical isomer, or salt of claim 19, wherein R3 is selected from the group consisting of 2-, 3-, or 4-trifluoromethylphenylcarbonyl, 2-, 3-, or A- trifluoromethylbenzylcarbonyl, 2-, 3-, or 4-trifluoromethylbenzyloxycarbonyl, 2-, 3-, or 4- trifiuoromethylphenylamido, 2-, 3-, or 4-trifluoromethylbenzylamido, and A-, 5-, 6-, or 7- trifluoromethyl- 1 H-benzimidazole-2-yl.
21. The compound, optical isomer, or salt of claim 19, wherein R is selected from the group consisting of 4-trifluoromethylphenylcarbonyl, 4-trifiuoromethylbenzylcarbonyl, A- trifluoromethylbenzyloxycarbonyl, 4-trifluoromethylphenylamido, A- trifluoromethylbenzylamido, 5 -trifluoromethyl- lH-benzimidazole-2-yl, and 6- trifluoromethyl- 1 H-benzimidazole-2-yl .
22. The compound, optical isomer, or salt of any of claims 12-18, wherein the compound is selected from the group consisting of
23. A compound of the formula (II) :
H-Tyr-D-Ala-Phe-Gly-Tyr-Pro — R4 — R5
(H) wherein
R4 is one or more amino acid residues; and
R5 is selected from the group consisting of aryl, arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, aralkylamido, and heteroaryl, wherein the aryl, heteroaryl, or the aryl part of arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more fluoro or trifluoromethyl groups;
an optical isomer thereof; or a pharmaceutically acceptable salt thereof.
24. The compound, optical isomer, or salt of claim 23, wherein R4 is one or more amino acid residues selected from the group consisting of glycinyl, alaninyl, valinyl, leucinyl, isoleucinyl, phenylalaninyl, asparaginyl, glutaminyl, tryptophanyl, prolinyl, serinyl, threoninyl, tyrosinyl, hydroxyprolinyl, cysteinyl, cystinyl, methioninyl, aspartyl, glutamyl, lysinyl, argininyl, and histidinyl.
25. The compound, optical isomer, or salt of claim 23 or 24, wherein R4 is one or more amino acid residues selected from the group consisting of phenylalaninyl, asparaginyl, glutaminyl, serinyl, and lysinyl.
26. The compound, optical isomer, or salt of any of claims 23-25, wherein the aryl, heteroaryl, or the aryl part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more fluoro groups.
27. The compound, optical isomer, or salt of claim 26, wherein R5 is selected from the group consisting of 2-, 3-, or 4-fluorophenylcarbonyl, 2-, 3-, or 4-fluorobenzylcarbonyl, 2-, 3-, or 4-fluorobenzyloxycarbonyl, 2-, 3-, or 4-fluorophenylamido, 2-, 3-, or A- fluorobenzylamido, and 4-, 5-, 6-, or 7-fluoro-lH-benzimidazole-2-yl.
28. The compound, optical isomer, or salt of claim 26, wherein R5 is selected from the group consisting of 4-fluorophenylcarbonyl, 4-fluorobenzylcarbonyl, A- fluorobenzyloxycarbonyl, 4-fluorophenylamido, 4-fluorobenzylamido, 5-fluoro-lH- benzimidazole-2-yl, and 6-fluoro-lH-benzimidazole-2-yl.
29. The compound, optical isomer, or salt of any of claims 23-25, wherein the aryl, heteroaryl, or the aryl part of arylcarbonyl, arylalkylcarbonyl, aralkyloxycarbonyl, arylamido, and aralkylamido is substituted with one or more trifluoromethyl groups
30. The compound, optical isomer, or salt of claim 29, wherein R5 is selected from the group consisting of 2-, 3-, or 4-trifluoromethylphenylcarbonyl, 2-, 3-, or A- trifluoromethylbenzylcarbonyl, 2-, 3-, or 4-trifluoromethylbenzyloxycarbonyl, 2-, 3-, or A- trifluoromethylphenylamido, 2-, 3-, or 4-trifluoromethylbenzylamido, and A-, 5-, 6-, or 7- trifluoromethyl- 1 H-benzimidazole-2-yl.
31. The compound, optical isomer, or salt of claim 29, wherein R5 is selected from the group consisting of 4-trifluoromethylphenylcarbonyl, 4-trifluoromethylbenzylcarbonyl, A- trifluoromethylbenzyloxycarbonyl, 4-trifluoromethylphenylamido, 4- trifluoromethylbenzylamido, 5-trifluoromethyl-lH-benzimidazole-2-yl, and 6- trifluoromethyl- 1 H-benzimidazole-2-yl .
32. The compound, optical isomer, or salt of any of claims 23-28, wherein the compound is selected from the group consisting of
(H)
wherein, the fluoro is at the 2, 3, or 4 position.
33. The compound, optical isomer, or salt of claim 32, wherein the fluoro is at the 4 position.
34. The compound, optical isomer, or salt of any of claims 1-33, wherein the fluoro is 18F.
35. A pharmaceutical composition comprising a compound, optical isomer, or salt of any of claims 1-35 and a pharmaceutically acceptable carrier.
36. A method of locating a μ- and/or δ-opioid receptor that is contained in a tissue or organ of a subject comprising
a) administering to the subject a radiolabeled compound, optical isomer, or salt of any of claims 1-34,
b) obtaining a diagnostic image of the tissue or organ, c) determining the location of radiolabeled compound bound to the tissue or organ, and
d) correlating the location of the bound radiolabeled compound with the location of the receptor in the subject.
37. A method of measuring the quantity of a μ- and/or δ-opioid receptor that is contained in a tissue or organ of a subject comprising
a) administering to the subject a radiolabeled compound, optical isomer, or salt of any of claims 1-34,
b) obtaining a positron emission tomography (PET) image of the tissue or organ,
c) determining the amount of radiolabeled compound bound to the tissue or organ, and
d) correlating the amount of the bound radiolabeled compound with the quantity of receptor in the subject.
38. The method of claim 36 or 37, wherein the receptor is a μ-opioid receptor.
39. The method of claim 36 or 37, wherein the receptor is a δ-opioid receptor.
40. The method of any of claims 30-33, wherein the radiolabeled compound, optical isomer, or salt is administered in an amount effective to provide an image.
41. The method of any of claims 36-39, wherein step b) comprises exposing the tissue or organ in the subject to an energy source, whereupon a diagnostic image of the tissue or organ is obtained.
42. The method of claim 36, wherein the diagnostic image is positron emission tomography (PET) image, a magnetic resonance image (MRI), a computerized tomography (CT) scan, an x-ray contrast image, single photon emission computed spectroscopy (SPECT) image, or a combination thereof.
43. The method of claim 36, wherein step b) comprises obtaining a positron emission tomography (PET) image of the tissue or organ.
44. Use of the compound , optical isomer, or salt of any one of claims 1 to 34 or the composition of claim 35, in the manufacture of an agent for locating a tissue comprising a μ- or δ-opioid receptor.
45. The use according to claim 44, wherein the tissue is selected from the group consisting of breast tissue, ovary tissue, salivary gland tissue, stomach tissue, kidney tissue, colon tissue, rectal tissue, cervical tissue, bladder tissue, head tissue, neck tissue, esophageal tissue, lung tissue, tracheal tissue, nasal tissue, mouth tissue, laryngeal tissue, pharyngeal tissue, and epiglotteal tissue.
46. The use according to claim 45, wherein the tissue is selected from the group consisting of lung tissue, colon tissue, rectal tissue, and breast tissue.
47. The use according to claim 46, wherein the tissue is lung tissue.
48. Use of the compound, optical isomer, or salt of any one of claims 1 to 34 or the composition of claim 35, in the manufacture of an agent for locating a cancer cell comprising a μ- or δ-opioid receptor.
49. The use according to claim 48, wherein the cancer cell is selected from the group consisting of NCI-H146, NCI-Hl 87, SHSY5Y, NS20Y, SK-N-SH, NG108-15, and T47D.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US97014307P | 2007-09-05 | 2007-09-05 | |
US60/970,143 | 2007-09-05 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2009032840A1 true WO2009032840A1 (en) | 2009-03-12 |
Family
ID=40239808
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2008/075114 WO2009032840A1 (en) | 2007-09-05 | 2008-09-03 | Fluorine- substituted 2 ', 6 ' -dimethyl-l-tyrosine-1, 2,3, 4-tetrahydr0is0quin0line-3-carb0xylic acid peptides (dmt-tic) for use as myu- and delta-opioid receptor probes in |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2009032840A1 (en) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN110194785A (en) * | 2016-04-21 | 2019-09-03 | 兰州大学 | The multiple target point peptides molecule and its preparation of a kind of opium and neuropeptide FF receptor and application |
EP3807296A4 (en) * | 2018-06-14 | 2022-03-02 | Dana-Farber Cancer Institute, Inc. | PEPTIDOMIMETIC INHIBITORS OF PEPTIDYL-PROLYL-CIS/TRANS-ISOMERASE (PIN1) |
-
2008
- 2008-09-03 WO PCT/US2008/075114 patent/WO2009032840A1/en active Application Filing
Non-Patent Citations (3)
Title |
---|
BALBONI ET AL: "Effect of Lysine at C-terminus of the Dmt-Tic Opioid Pharmacophore", JOURNAL OF MEDICINAL CHEMISTRY, vol. 49, no. 18, 16 August 2006 (2006-08-16), pages 5610 - 5617, XP002511398 * |
RAVERT ET AL: "PET imaging of opioid receptors in pain: progress and new directions", CURRENT PHARMACEUTICAL DESIGN, vol. 10, no. 7, 2004, XP002511399 * |
RYU ET AL: "Synthesis of a Potent and Selective 18F-Labeled ?-Opioid Receptor Antagonist Derived from the Dmt-Tic Pharmacophore for Positron Emission Tomography Imaging", JOURNAL OF MEDICINAL CHEMISTRY, vol. 51, no. 6, 1 March 2008 (2008-03-01), pages 1817 - 1823, XP002511400 * |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN110194785A (en) * | 2016-04-21 | 2019-09-03 | 兰州大学 | The multiple target point peptides molecule and its preparation of a kind of opium and neuropeptide FF receptor and application |
CN110194785B (en) * | 2016-04-21 | 2023-02-24 | 上海天慈生命科学发展有限公司 | Multi-target peptide molecule of opioid and neuropeptide FF receptor and preparation and application thereof |
EP3807296A4 (en) * | 2018-06-14 | 2022-03-02 | Dana-Farber Cancer Institute, Inc. | PEPTIDOMIMETIC INHIBITORS OF PEPTIDYL-PROLYL-CIS/TRANS-ISOMERASE (PIN1) |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
KR101319740B1 (en) | Cyclic peptide cxcr4 antagonists | |
JP6576828B2 (en) | Neurotensin receptor ligand | |
US8628750B2 (en) | Cancer imaging and treatment | |
US11992536B2 (en) | Dual-targeting compound and preparation method and application thereof | |
EP3152226B1 (en) | Modified cyclopentapeptides and uses thereof | |
EP0644772B1 (en) | Opioid peptides | |
US9266924B2 (en) | Cyclopentapeptide derivatives and uses thereof | |
JPH11503420A (en) | Radiolabeled peptides for diagnostics and therapeutics | |
AU2019469641B2 (en) | Radiolabeled bombesin-derived compounds for in vivo imaging of gastrin-releasing peptide receptor (GRPR) and treatment of GRPR-related disorders | |
CN111065646A (en) | radiopharmaceutical | |
US11160887B2 (en) | Compositions for use in diagnosing and treating melanoma, including metastatic melanoma and methods related to same | |
WO2023164775A1 (en) | Radiolabeled compounds targetng the prostate-specific membrane antigen | |
WO2009032840A1 (en) | Fluorine- substituted 2 ', 6 ' -dimethyl-l-tyrosine-1, 2,3, 4-tetrahydr0is0quin0line-3-carb0xylic acid peptides (dmt-tic) for use as myu- and delta-opioid receptor probes in | |
WO2011131731A1 (en) | Cyclopeptide derivatives and uses thereof | |
EP1257575B1 (en) | Radiolabeled vasoactive intestinal peptide analogs for diagnosis and radiotherapy | |
US8512675B2 (en) | N and/or Nα derivatized, metal and organic protected L-histidine for coupling to biomolecules for highly efficient labeling with [M(OH2)3 (CO)3]+ by fac coordination | |
RU2838179C2 (en) | Dual targeting compound, method for its preparation and use | |
EP0498771A2 (en) | MSH peptide derivatives | |
Sasaki et al. | Synthesis of a potent and selective 18F-labeled δ-opioid receptor antagonist derived from the Dmt-Tic pharmacophore for PET imaging | |
WO2023201435A1 (en) | Cxcr4-targeting compounds, and methods of making and using the same | |
WO2024046469A1 (en) | Cyclic peptide and preparation method therefor, and complex comprising same and use thereof | |
ES2367941T3 (en) | CXCR4 PEPTIDE CYCLIC ANTAGONISTS. | |
RU2824959C2 (en) | Bicyclic peptide ligands specific for mt1-mmp | |
JP2005068046A (en) | Somatostatin analog derivatives and uses thereof | |
JPH06263798A (en) | Cyclic pentapeptide containing beta turn and gamma turn, its production and use thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 08829409 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 08829409 Country of ref document: EP Kind code of ref document: A1 |