NL2019752B1 - Cytotoxic Complexes - Google Patents
Cytotoxic Complexes Download PDFInfo
- Publication number
- NL2019752B1 NL2019752B1 NL2019752A NL2019752A NL2019752B1 NL 2019752 B1 NL2019752 B1 NL 2019752B1 NL 2019752 A NL2019752 A NL 2019752A NL 2019752 A NL2019752 A NL 2019752A NL 2019752 B1 NL2019752 B1 NL 2019752B1
- Authority
- NL
- Netherlands
- Prior art keywords
- cancer
- optionally substituted
- groups
- group
- compound
- Prior art date
Links
- 230000001472 cytotoxic effect Effects 0.000 title description 10
- 231100000433 cytotoxic Toxicity 0.000 title description 8
- KDLHZDBZIXYQEI-UHFFFAOYSA-N Palladium Chemical compound [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 claims abstract description 60
- 150000001875 compounds Chemical class 0.000 claims abstract description 57
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 claims abstract description 50
- 239000003446 ligand Substances 0.000 claims abstract description 43
- 229910052751 metal Inorganic materials 0.000 claims abstract description 27
- 239000000203 mixture Substances 0.000 claims abstract description 27
- 239000002184 metal Substances 0.000 claims abstract description 26
- 229910052763 palladium Inorganic materials 0.000 claims abstract description 18
- 229910052697 platinum Inorganic materials 0.000 claims abstract description 17
- 239000010931 gold Substances 0.000 claims abstract description 11
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 claims abstract description 8
- 229910052737 gold Inorganic materials 0.000 claims abstract description 8
- 150000001768 cations Chemical class 0.000 claims abstract description 3
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 claims description 33
- 206010028980 Neoplasm Diseases 0.000 claims description 20
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 claims description 18
- 201000011510 cancer Diseases 0.000 claims description 18
- 125000004076 pyridyl group Chemical group 0.000 claims description 16
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 claims description 15
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 claims description 12
- 238000000338 in vitro Methods 0.000 claims description 12
- -1 or good Substances 0.000 claims description 12
- 238000000034 method Methods 0.000 claims description 11
- 238000011282 treatment Methods 0.000 claims description 11
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 claims description 10
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 claims description 9
- 239000002246 antineoplastic agent Substances 0.000 claims description 9
- 239000003814 drug Substances 0.000 claims description 9
- 125000001033 ether group Chemical group 0.000 claims description 9
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 9
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 claims description 8
- 201000010099 disease Diseases 0.000 claims description 8
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 8
- 125000005843 halogen group Chemical group 0.000 claims description 8
- 150000003839 salts Chemical class 0.000 claims description 8
- 239000002904 solvent Substances 0.000 claims description 8
- 125000000547 substituted alkyl group Chemical group 0.000 claims description 8
- 125000003903 2-propenyl group Chemical group [H]C([*])([H])C([H])=C([H])[H] 0.000 claims description 6
- 125000004185 ester group Chemical group 0.000 claims description 6
- 229910052757 nitrogen Inorganic materials 0.000 claims description 6
- 125000003107 substituted aryl group Chemical group 0.000 claims description 6
- 206010005003 Bladder cancer Diseases 0.000 claims description 5
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 claims description 5
- 206010033128 Ovarian cancer Diseases 0.000 claims description 5
- 206010061535 Ovarian neoplasm Diseases 0.000 claims description 5
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 claims description 5
- 150000001450 anions Chemical class 0.000 claims description 5
- ITMCEJHCFYSIIV-UHFFFAOYSA-M triflate Chemical compound [O-]S(=O)(=O)C(F)(F)F ITMCEJHCFYSIIV-UHFFFAOYSA-M 0.000 claims description 5
- 201000005112 urinary bladder cancer Diseases 0.000 claims description 5
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 4
- NHNBFGGVMKEFGY-UHFFFAOYSA-N Nitrate Chemical compound [O-][N+]([O-])=O NHNBFGGVMKEFGY-UHFFFAOYSA-N 0.000 claims description 4
- 208000024313 Testicular Neoplasms Diseases 0.000 claims description 4
- 206010057644 Testis cancer Diseases 0.000 claims description 4
- DTQVDTLACAAQTR-UHFFFAOYSA-M Trifluoroacetate Chemical compound [O-]C(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-M 0.000 claims description 4
- 125000003277 amino group Chemical group 0.000 claims description 4
- 230000012010 growth Effects 0.000 claims description 4
- 208000015181 infectious disease Diseases 0.000 claims description 4
- 201000005202 lung cancer Diseases 0.000 claims description 4
- 208000020816 lung neoplasm Diseases 0.000 claims description 4
- 208000016691 refractory malignant neoplasm Diseases 0.000 claims description 4
- 201000003120 testicular cancer Diseases 0.000 claims description 4
- 230000004222 uncontrolled growth Effects 0.000 claims description 4
- CPELXLSAUQHCOX-UHFFFAOYSA-M Bromide Chemical compound [Br-] CPELXLSAUQHCOX-UHFFFAOYSA-M 0.000 claims description 3
- 206010009944 Colon cancer Diseases 0.000 claims description 3
- KRHYYFGTRYWZRS-UHFFFAOYSA-M Fluoride anion Chemical compound [F-] KRHYYFGTRYWZRS-UHFFFAOYSA-M 0.000 claims description 3
- 229910019142 PO4 Inorganic materials 0.000 claims description 3
- 208000000453 Skin Neoplasms Diseases 0.000 claims description 3
- QAOWNCQODCNURD-UHFFFAOYSA-L Sulfate Chemical compound [O-]S([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 claims description 3
- 125000003118 aryl group Chemical group 0.000 claims description 3
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 claims description 3
- 230000010261 cell growth Effects 0.000 claims description 3
- 208000029742 colonic neoplasm Diseases 0.000 claims description 3
- XMBWDFGMSWQBCA-UHFFFAOYSA-N hydrogen iodide Chemical compound I XMBWDFGMSWQBCA-UHFFFAOYSA-N 0.000 claims description 3
- 125000001570 methylene group Chemical group [H]C([H])([*:1])[*:2] 0.000 claims description 3
- 125000002950 monocyclic group Chemical group 0.000 claims description 3
- 125000004430 oxygen atom Chemical group O* 0.000 claims description 3
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 claims description 3
- 239000010452 phosphate Substances 0.000 claims description 3
- 230000000979 retarding effect Effects 0.000 claims description 3
- 201000000849 skin cancer Diseases 0.000 claims description 3
- 125000004962 sulfoxyl group Chemical group 0.000 claims description 3
- ZXMGHDIOOHOAAE-UHFFFAOYSA-N 1,1,1-trifluoro-n-(trifluoromethylsulfonyl)methanesulfonamide Chemical compound FC(F)(F)S(=O)(=O)NS(=O)(=O)C(F)(F)F ZXMGHDIOOHOAAE-UHFFFAOYSA-N 0.000 claims description 2
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 claims description 2
- IOVCWXUNBOPUCH-UHFFFAOYSA-M Nitrite anion Chemical compound [O-]N=O IOVCWXUNBOPUCH-UHFFFAOYSA-M 0.000 claims description 2
- 239000002671 adjuvant Substances 0.000 claims description 2
- 239000011203 carbon fibre reinforced carbon Substances 0.000 claims description 2
- 150000001732 carboxylic acid derivatives Chemical class 0.000 claims description 2
- 229940127089 cytotoxic agent Drugs 0.000 claims description 2
- 239000002254 cytotoxic agent Substances 0.000 claims description 2
- 231100000599 cytotoxic agent Toxicity 0.000 claims description 2
- 239000003085 diluting agent Substances 0.000 claims description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-M hydroxide Chemical compound [OH-] XLYOFNOQVPJJNP-UHFFFAOYSA-M 0.000 claims description 2
- 229940005654 nitrite ion Drugs 0.000 claims description 2
- YPJUNDFVDDCYIH-UHFFFAOYSA-N perfluorobutyric acid Chemical compound OC(=O)C(F)(F)C(F)(F)C(F)(F)F YPJUNDFVDDCYIH-UHFFFAOYSA-N 0.000 claims description 2
- 239000008194 pharmaceutical composition Substances 0.000 claims description 2
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 2
- JOXIMZWYDAKGHI-UHFFFAOYSA-N toluene-4-sulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-N 0.000 claims description 2
- AFVFQIVMOAPDHO-UHFFFAOYSA-M Methanesulfonate Chemical compound CS([O-])(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-M 0.000 claims 1
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 claims 1
- 239000003795 chemical substances by application Substances 0.000 claims 1
- MUJIDPITZJWBSW-UHFFFAOYSA-N palladium(2+) Chemical compound [Pd+2] MUJIDPITZJWBSW-UHFFFAOYSA-N 0.000 claims 1
- HRGDZIGMBDGFTC-UHFFFAOYSA-N platinum(2+) Chemical compound [Pt+2] HRGDZIGMBDGFTC-UHFFFAOYSA-N 0.000 claims 1
- 229910052717 sulfur Inorganic materials 0.000 claims 1
- 125000004434 sulfur atom Chemical group 0.000 claims 1
- 210000001550 testis Anatomy 0.000 claims 1
- 230000001225 therapeutic effect Effects 0.000 claims 1
- 125000004424 polypyridyl Polymers 0.000 abstract description 3
- 210000004027 cell Anatomy 0.000 description 58
- 231100000135 cytotoxicity Toxicity 0.000 description 20
- 230000003013 cytotoxicity Effects 0.000 description 19
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 17
- 229960004316 cisplatin Drugs 0.000 description 17
- 230000015572 biosynthetic process Effects 0.000 description 15
- 239000000725 suspension Substances 0.000 description 15
- 239000000243 solution Substances 0.000 description 14
- 230000000694 effects Effects 0.000 description 13
- 238000003786 synthesis reaction Methods 0.000 description 12
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 9
- 238000001727 in vivo Methods 0.000 description 8
- 230000004700 cellular uptake Effects 0.000 description 7
- 229940079593 drug Drugs 0.000 description 7
- 239000000843 powder Substances 0.000 description 7
- 230000001093 anti-cancer Effects 0.000 description 6
- 239000013078 crystal Substances 0.000 description 6
- 229940041181 antineoplastic drug Drugs 0.000 description 5
- 230000005595 deprotonation Effects 0.000 description 5
- 238000010537 deprotonation reaction Methods 0.000 description 5
- IDGUHHHQCWSQLU-UHFFFAOYSA-N ethanol;hydrate Chemical compound O.CCO IDGUHHHQCWSQLU-UHFFFAOYSA-N 0.000 description 5
- 230000010534 mechanism of action Effects 0.000 description 5
- 239000012528 membrane Substances 0.000 description 5
- 229920006395 saturated elastomer Polymers 0.000 description 5
- 239000007787 solid Substances 0.000 description 5
- 241000699670 Mus sp. Species 0.000 description 4
- 238000005481 NMR spectroscopy Methods 0.000 description 4
- 230000002378 acidificating effect Effects 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 239000000706 filtrate Substances 0.000 description 4
- 230000003993 interaction Effects 0.000 description 4
- 239000000523 sample Substances 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- KBPLFHHGFOOTCA-UHFFFAOYSA-N 1-Octanol Chemical class CCCCCCCCO KBPLFHHGFOOTCA-UHFFFAOYSA-N 0.000 description 3
- GRYLNZFGIOXLOG-UHFFFAOYSA-N Nitric acid Chemical compound O[N+]([O-])=O GRYLNZFGIOXLOG-UHFFFAOYSA-N 0.000 description 3
- 238000002835 absorbance Methods 0.000 description 3
- 238000000862 absorption spectrum Methods 0.000 description 3
- 150000001412 amines Chemical class 0.000 description 3
- 239000007864 aqueous solution Substances 0.000 description 3
- 230000004071 biological effect Effects 0.000 description 3
- 238000002474 experimental method Methods 0.000 description 3
- 230000001965 increasing effect Effects 0.000 description 3
- 210000004072 lung Anatomy 0.000 description 3
- 238000004519 manufacturing process Methods 0.000 description 3
- 229910021645 metal ion Inorganic materials 0.000 description 3
- 229910017604 nitric acid Inorganic materials 0.000 description 3
- 239000012299 nitrogen atmosphere Substances 0.000 description 3
- 150000002940 palladium Chemical class 0.000 description 3
- 150000002941 palladium compounds Chemical class 0.000 description 3
- 150000003057 platinum Chemical class 0.000 description 3
- 239000002244 precipitate Substances 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 238000001542 size-exclusion chromatography Methods 0.000 description 3
- 241000894007 species Species 0.000 description 3
- 210000004881 tumor cell Anatomy 0.000 description 3
- 230000004614 tumor growth Effects 0.000 description 3
- IOOMXAQUNPWDLL-UHFFFAOYSA-N 2-[6-(diethylamino)-3-(diethyliminiumyl)-3h-xanthen-9-yl]-5-sulfobenzene-1-sulfonate Chemical compound C=12C=CC(=[N+](CC)CC)C=C2OC2=CC(N(CC)CC)=CC=C2C=1C1=CC=C(S(O)(=O)=O)C=C1S([O-])(=O)=O IOOMXAQUNPWDLL-UHFFFAOYSA-N 0.000 description 2
- ROFVEXUMMXZLPA-UHFFFAOYSA-N Bipyridyl Chemical group N1=CC=CC=C1C1=CC=CC=N1 ROFVEXUMMXZLPA-UHFFFAOYSA-N 0.000 description 2
- OKKJLVBELUTLKV-MZCSYVLQSA-N Deuterated methanol Chemical compound [2H]OC([2H])([2H])[2H] OKKJLVBELUTLKV-MZCSYVLQSA-N 0.000 description 2
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 2
- 229910002651 NO3 Inorganic materials 0.000 description 2
- PXHVJJICTQNCMI-UHFFFAOYSA-N Nickel Chemical compound [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 description 2
- 229920005654 Sephadex Polymers 0.000 description 2
- 239000012507 Sephadex™ Substances 0.000 description 2
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical group [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 2
- 238000010521 absorption reaction Methods 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 150000003841 chloride salts Chemical class 0.000 description 2
- 150000004696 coordination complex Chemical class 0.000 description 2
- 125000004122 cyclic group Chemical group 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 238000009792 diffusion process Methods 0.000 description 2
- 239000003480 eluent Substances 0.000 description 2
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 238000001095 inductively coupled plasma mass spectrometry Methods 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 230000003834 intracellular effect Effects 0.000 description 2
- 150000002500 ions Chemical class 0.000 description 2
- 210000005265 lung cell Anatomy 0.000 description 2
- 230000003211 malignant effect Effects 0.000 description 2
- 239000002609 medium Substances 0.000 description 2
- 238000001139 pH measurement Methods 0.000 description 2
- VLTRZXGMWDSKGL-UHFFFAOYSA-M perchlorate Inorganic materials [O-]Cl(=O)(=O)=O VLTRZXGMWDSKGL-UHFFFAOYSA-M 0.000 description 2
- VLTRZXGMWDSKGL-UHFFFAOYSA-N perchloric acid Chemical compound OCl(=O)(=O)=O VLTRZXGMWDSKGL-UHFFFAOYSA-N 0.000 description 2
- 230000000737 periodic effect Effects 0.000 description 2
- 238000001556 precipitation Methods 0.000 description 2
- 239000000047 product Substances 0.000 description 2
- 230000002062 proliferating effect Effects 0.000 description 2
- 102000004169 proteins and genes Human genes 0.000 description 2
- 108090000623 proteins and genes Proteins 0.000 description 2
- 239000003642 reactive oxygen metabolite Substances 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 238000002390 rotary evaporation Methods 0.000 description 2
- 238000012360 testing method Methods 0.000 description 2
- 238000004448 titration Methods 0.000 description 2
- 229910052723 transition metal Inorganic materials 0.000 description 2
- 150000003624 transition metals Chemical class 0.000 description 2
- 238000002371 ultraviolet--visible spectrum Methods 0.000 description 2
- 238000001644 13C nuclear magnetic resonance spectroscopy Methods 0.000 description 1
- 238000005160 1H NMR spectroscopy Methods 0.000 description 1
- STQGQHZAVUOBTE-UHFFFAOYSA-N 7-Cyan-hept-2t-en-4,6-diinsaeure Natural products C1=2C(O)=C3C(=O)C=4C(OC)=CC=CC=4C(=O)C3=C(O)C=2CC(O)(C(C)=O)CC1OC1CC(N)C(O)C(C)O1 STQGQHZAVUOBTE-UHFFFAOYSA-N 0.000 description 1
- 238000011725 BALB/c mouse Methods 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- 230000005778 DNA damage Effects 0.000 description 1
- 231100000277 DNA damage Toxicity 0.000 description 1
- 108010024636 Glutathione Proteins 0.000 description 1
- 101000666657 Homo sapiens Rho-related GTP-binding protein RhoQ Proteins 0.000 description 1
- 238000012404 In vitro experiment Methods 0.000 description 1
- 229910021135 KPF6 Inorganic materials 0.000 description 1
- 206010028813 Nausea Diseases 0.000 description 1
- 206010029155 Nephropathy toxic Diseases 0.000 description 1
- 206010029350 Neurotoxicity Diseases 0.000 description 1
- 239000012124 Opti-MEM Substances 0.000 description 1
- 206010033109 Ototoxicity Diseases 0.000 description 1
- 102100038339 Rho-related GTP-binding protein RhoQ Human genes 0.000 description 1
- 238000004639 Schlenk technique Methods 0.000 description 1
- 206010044221 Toxic encephalopathy Diseases 0.000 description 1
- 238000002441 X-ray diffraction Methods 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 239000008346 aqueous phase Substances 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 125000002843 carboxylic acid group Chemical group 0.000 description 1
- 239000003054 catalyst Substances 0.000 description 1
- 125000002091 cationic group Chemical group 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 230000003833 cell viability Effects 0.000 description 1
- 230000005889 cellular cytotoxicity Effects 0.000 description 1
- 230000010001 cellular homeostasis Effects 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- PBAYDYUZOSNJGU-UHFFFAOYSA-N chelidonic acid Natural products OC(=O)C1=CC(=O)C=C(C(O)=O)O1 PBAYDYUZOSNJGU-UHFFFAOYSA-N 0.000 description 1
- 230000000973 chemotherapeutic effect Effects 0.000 description 1
- 150000001805 chlorine compounds Chemical class 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 238000012258 culturing Methods 0.000 description 1
- 238000011461 current therapy Methods 0.000 description 1
- 238000002784 cytotoxicity assay Methods 0.000 description 1
- 231100000263 cytotoxicity test Toxicity 0.000 description 1
- STQGQHZAVUOBTE-VGBVRHCVSA-N daunorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 description 1
- 229960000975 daunorubicin Drugs 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 238000001784 detoxification Methods 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 238000006073 displacement reaction Methods 0.000 description 1
- 238000000921 elemental analysis Methods 0.000 description 1
- 230000002349 favourable effect Effects 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 238000005194 fractionation Methods 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 229960003180 glutathione Drugs 0.000 description 1
- CBMIPXHVOVTTTL-UHFFFAOYSA-N gold(3+) Chemical compound [Au+3] CBMIPXHVOVTTTL-UHFFFAOYSA-N 0.000 description 1
- 238000004896 high resolution mass spectrometry Methods 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 229910052738 indium Inorganic materials 0.000 description 1
- APFVFJFRJDLVQX-UHFFFAOYSA-N indium atom Chemical compound [In] APFVFJFRJDLVQX-UHFFFAOYSA-N 0.000 description 1
- 238000002354 inductively-coupled plasma atomic emission spectroscopy Methods 0.000 description 1
- 238000009830 intercalation Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 150000002505 iron Chemical class 0.000 description 1
- 230000002427 irreversible effect Effects 0.000 description 1
- 125000002183 isoquinolinyl group Chemical group C1(=NC=CC2=CC=CC=C12)* 0.000 description 1
- 238000001819 mass spectrum Methods 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 231100000682 maximum tolerated dose Toxicity 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- JZMJDSHXVKJFKW-UHFFFAOYSA-M methyl sulfate(1-) Chemical compound COS([O-])(=O)=O JZMJDSHXVKJFKW-UHFFFAOYSA-M 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- COTLTZSJWUVSPY-UHFFFAOYSA-N n-pyridin-2-yl-6-[6-(pyridin-2-ylamino)pyridin-2-yl]pyridin-2-amine Chemical compound C=1C=CC(C=2N=C(NC=3N=CC=CC=3)C=CC=2)=NC=1NC1=CC=CC=N1 COTLTZSJWUVSPY-UHFFFAOYSA-N 0.000 description 1
- 230000008693 nausea Effects 0.000 description 1
- 231100000417 nephrotoxicity Toxicity 0.000 description 1
- 230000007694 nephrotoxicity Effects 0.000 description 1
- 231100000228 neurotoxicity Toxicity 0.000 description 1
- 230000007135 neurotoxicity Effects 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 229910052759 nickel Inorganic materials 0.000 description 1
- 125000004433 nitrogen atom Chemical group N* 0.000 description 1
- 238000000655 nuclear magnetic resonance spectrum Methods 0.000 description 1
- 238000010899 nucleation Methods 0.000 description 1
- 231100000262 ototoxicity Toxicity 0.000 description 1
- JRTYPQGPARWINR-UHFFFAOYSA-N palladium platinum Chemical compound [Pd].[Pt] JRTYPQGPARWINR-UHFFFAOYSA-N 0.000 description 1
- 238000005192 partition Methods 0.000 description 1
- 239000012071 phase Substances 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- NDBYXKQCPYUOMI-UHFFFAOYSA-N platinum(4+) Chemical compound [Pt+4] NDBYXKQCPYUOMI-UHFFFAOYSA-N 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 229940002612 prodrug Drugs 0.000 description 1
- 239000000651 prodrug Substances 0.000 description 1
- 230000005588 protonation Effects 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 150000003222 pyridines Chemical class 0.000 description 1
- LISFMEBWQUVKPJ-UHFFFAOYSA-N quinolin-2-ol Chemical compound C1=CC=C2NC(=O)C=CC2=C1 LISFMEBWQUVKPJ-UHFFFAOYSA-N 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 239000013074 reference sample Substances 0.000 description 1
- 150000003303 ruthenium Chemical class 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 229910052709 silver Inorganic materials 0.000 description 1
- 239000004332 silver Substances 0.000 description 1
- 238000005063 solubilization Methods 0.000 description 1
- 230000007928 solubilization Effects 0.000 description 1
- 239000012453 solvate Substances 0.000 description 1
- 238000003756 stirring Methods 0.000 description 1
- 239000011550 stock solution Substances 0.000 description 1
- 125000001424 substituent group Chemical group 0.000 description 1
- 229910021653 sulphate ion Inorganic materials 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 150000003573 thiols Chemical class 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 238000002627 tracheal intubation Methods 0.000 description 1
- 238000007492 two-way ANOVA Methods 0.000 description 1
- 238000000870 ultraviolet spectroscopy Methods 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07F—ACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
- C07F1/00—Compounds containing elements of Groups 1 or 11 of the Periodic Table
- C07F1/12—Gold compounds
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D213/00—Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
- C07D213/02—Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
- C07D213/04—Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
- C07D213/60—Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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 ring carbon atoms
- C07D213/72—Nitrogen atoms
- C07D213/74—Amino or imino radicals substituted by hydrocarbon or substituted hydrocarbon radicals
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/14—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
- C07D471/04—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07F—ACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
- C07F15/00—Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table
- C07F15/0006—Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table compounds of the platinum group
- C07F15/006—Palladium compounds
- C07F15/0066—Palladium compounds without a metal-carbon linkage
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07F—ACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
- C07F15/00—Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table
- C07F15/0006—Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table compounds of the platinum group
- C07F15/0086—Platinum compounds
- C07F15/0093—Platinum compounds without a metal-carbon linkage
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention relates to a compound of the formula(|): [Z—L]MQn (I) wherein Z represents a metal cation selected from the group consisting of palladium, platinum or gold or mixtures thereof; L is a tetradentate polypyridyl ligand ofthe general formula N-N’-N’-N (II) bonded to to Z to form a complex Z-L: n \ _ Hr \ m x ï W / H H N— \ H — H R (II).
Description
Cytotoxic Complexes
The present invention relates to novel metallo tetrapyridyl complexes for use as cytotoxic anti-cancer or antibacterial agents, and to their use in the preparation of medication for the treatment of unlimited cell growth, such as in infections or in cancer and tumour cells, and their preparation.
Background of the Invention
Current therapies directed against cells with uncontrolled growth properties, such as highly proliferating cells, cancerous cells, and tumour cells, require that they include a set of unique criteria in order to avoid damaging normal growing and/or non-proliferating cells.
The leading anticancer drug cisplatin is one of the landmarks in modern inorganic chemistry. In the body, cisplatin enters cells, hydrolyses, and forms irreversible adducts with DNA, ultimately triggering cell death via apoptosis. However, because of their lack of selectivity, severe side effects are commonly associated with the use of cisplatin, including nephrotoxicity, neurotoxicity, ototoxicity, and nausea. Furthermore, cancer cells can acquire increasing levels of cisplatin-resistance. Accordingly, new anticancer metallodrugs that may offer less severe side effects, and may retain activity against resistant cells would be highly desirable.
Summary of the Invention
Accordingly, in a first aspect, the present invention relates to a metallo tetrapyridyl ligand complexes based on periodic system column 10 or 11 transition metals according to claim 1, which were found to have chemotherapeutic activities, particularly anticancer properties. In more detail, the present invention relates to a compound of the formula(l):
wherein Z represents a metal cation selected from the group consisting of palladium, platinum or gold or mixtures thereof; L is a tetradentate polypyridyl ligand of the general formula N-N'-N'-N (II) bonded to to Z to form a complex Z-L:
wherein group X may independently represent a bridging group connecting two adjacent pyridyl groups, and may be an optionally substituted amino group, an optionally substituted methylene group, carboxyl or sulfoxyl group, a sulphur atom, or an oxygen atom; wherein group Y may be as group X, or may independently represent two hydrogen atoms, halogen atoms, optionally substituted alkyl groups, optionally substituted aryl groups, optionally substituted allyl groups, optionally substituted ester groups, optionally substituted ether groups; M is an integer representing the positive charge of the Z-L complex; Q represents the eounter-anion(s) ionically bonded to the Z-L complex; and n is an integer representing the number of counter anions ionically bonded to the Z-L complex; and R, R', R" and R"' independently represent one or more hydrogen atoms, halogen atoms, hydroxide, carboxylic acid, optionally substituted alkyl groups, optionally substituted aryl groups, optionally substituted allyl groups, optionally substituted ester groups, optionally substituted ether groups, or R, R', R" and R'" represents a monocyclic or bicyclic aliphatic or aromatic structure with one pyridyl groups, or two groups R, R', R" and R'" may together form a tricyclic structure with two adjacent pyridyl groups, and
Riv and Rv may represent a carbon-carbon bridge between two adjacent pyridyl groups, or in the case wherein X and V are both amino groups linking two pyridyl groups, may each individually represent a hydrogen atom, a halogen atom, an optionally substituted alkyl group, and an optionally substituted ether group.
In a second aspect, the present invention also relates to the synthesis of a series of new metallo complexes based on tetrapyridyl ligands coordinated to periodic system column 10 or 11 transition metals, in particular gold (III), palladium(ll), and platinum(ll). Applicants surprisingly found that the in vitro cytotoxic activity of these complexes ranges from EC50 values in micromolar concentrations to spectacular nanomolar EC50 values.
In a further aspect, the present invention relates to the preparation of these compounds, their biochemical activity in vitro and in vivo, and to the use of the complexes in the manufacture of a medication for the treatment of unlimited cell growth, such as in infections, in cancer and/or tumour cells.
Influence of pH, interactions with DNA, cellular uptake, cellular fractionation, cell cytotoxicity on several commercially available human cancer cell lines and 1 non-cancerous cell line, in vitro mechanism of action, and in vivo studies have been performed.
Brief Description of the Figures
Fig. 1 discloses the chemical structures of a number of suitable ligands.
Figure 2 discloses the chemical structures of two tetrapyridyl ligands H2bapbpy, and Hbbpya.
Figure 3 shows a schematic synthesis of [l]Ch and [2]Ch
Figure 4 shows a schematic synthesis of [3]PF6 and [4] PFe, wherein L represents two adiditonal counterions PFe
Figure 5 shows palladium(ll) and platinum(ll) complexes of H2bapbpy (N-(6-(6-(pyridin- 2-ylamino)pyridin-2-yl)pyridin-2-yl)pyridin-2-amine) and Hbbpya (N,N-bis(2,2'-bipyrid-6-yl)amine). Hbbpya is a tetrapyridyl ligand similar to H2bapbpy except for its single non-coordinated amine bridge (while H2bapbpy has two).
Figure 6 shows the displacement ellipsoid of the cationic part of [1]CI2 (50% probability level) and of [Au(bapbpy)]PF6. Counter anions and lattice solvent molecules have been omitted for clarity.
Figure 7 illustrates the evolution of the UV-vis spectrum of [1]2+ (left) and [2]2+ (right) upon increasing the pH of an HCI aqueous solution (33 mM) of the metal complex using NaOH. [Pd] = [Pt] = 67 μΜ.
Figure 8: depicts the tumour volume of Balb/c mice treated with [3](PF6)2 and [4](PF6)2. CT-26 cells were injected subcutaneously into the right flank of Balb/c mice. Mice were treated on day 3, 5, 7,10, and 12 (indicated by black triangles) with either compound [3](PF6)2 at concentrations of 2.5 mg/kg (i.p.) or [4](PF6)2 at concentrations of 2.8 mg/kg (i p) ***^ p <0.001, two-way ANOVA, Bonferroni post-test.
Detailed description of the invention
Inorganic anticancer compounds typically interact with biomolecules following two different modes. A first mode may be direct binding of biomolecules such as DNA or proteins to the metal center to form a metal-ligand coordination bond. The formation of these coordination bonds is commonly preceded by the release of a ligand present in the original or intermediate complex. For instance the anticancer drug cisplatin and most of its derivatives rely on this mechanism of action.
The second type of interaction may be an indirect binding mode of the complex with its biomolecular target. Typically, one of the ligands of a metal complex intercalates via π-π or hydrophobic interactions to DNA or proteins. Typical intercalators consist of planar polyaromatic and extended polypyridyl ligands. Whereas organic intercalators such as daunorubicin are widely used in the clinics as cancer-treatment, no inorganic intercalators have been employed clinically so far.
The present invention relates to the use of coordinatively saturated tetrapyridyl metal complexes as potentially intercalating anticancer drugs. Hence, the present invention relates to complexes with a variation of the metal centre selected from the group consisting of palladium(ll), platinum(ll), or gold(lll); and preferably, (ii) the use of palladium(IV) or platinum(IV) centers to facilitate a prodrug (photo)reduction strategy; or (iii) the use of cyclometallated versions of the H2bapbpy and Hbbpya ligands. Metal ion Z is centrally bonded to the ligand, thus forming a complex. N-N'-N'-N represents a tetradentate ligand. M represents the charge of the complex, and typically M=+2, but upon deprotonation of the NH bridging amines it can become lower by one (M=+l) or two (M=0) units. The number of counter anions will depend on the charge M of the complex and its protonation state. Solvates of these compounds, for example with acetonitrile, water, methanol, ethanol, or other organic molecules, are also included.
In the compound, group X may independently represent a bridging group connecting two adjacent pyridyl groups, and may be an optionally substituted amino group, an optionally substituted methylene group, carboxyl or sulfoxyl group, a sulphur atom, or an oxygen atom.
Group Y may be as X, thereby leading to an open circular ligand structure, or it may independently represent two hydrogen atoms, halogen atoms, optionally substituted alkyl groups, optionally substituted aryl groups, optionally substituted allyl groups, optionally substituted ester groups, optionally substituted ether groups, thereby leading to an embracing, non-cyclic ligand. R, R', R" and R'" independently represent one or more hydrogen atoms, halogen atoms, preferably chloride, bromide or fluoride, optionally substituted alkyl groups, optionally substituted aryl groups, optionally substituted allyl groups, optionally substituted ester groups, optionally substituted ether groups, hydroxyl groups, carboxylic acid groups, amines, or R represents a monocyclic or bicyclic aliphatic or aromatic structure with one pyridyl groups, or two groups R, R', R" and R'" may together form a tricyclic structure with two adjacent pyridyl groups, e.g. forming quinolone or isoquinoline structures. Preferably, there are only the necessary number of substituents on the pyridyl rings, e.g. chlorides or ether groups, which may vary the electronic structure and hence binding strength of the ligand to the metal ion.
Generally, suitable tetradentate ligands N-N'-N'-N typically comprise a good metal chelating ligand, and include, but are not limited to, the compounds disclosed in Figure 1. Q represents a counter-anion, for example, chloride or hexafluorophosphate, but it could also be triflate, iodide, perchlorate, nitrate, sulfate, phosphate, or other as set out below. The counter-anion of the complex may be any useful counter ion that allows formation of the complex, as well as use in the in vitro or in vivo processes. Preferably, Q is selected from the group consisting of fluoride, chloride, bromide, iodide, sulphate, methyl sulphate, tetrafluoroborate, hexafluorophosphate, tetraphenylborate, triflate (CFsSCV), nonaflate (CF3(CF2)sS02*), bis(triflyl)amide ((CFsSChhN"), trifluoroacetate (CF3CO2 ~), heptafluorobutanoate (CF3(CF2)3C02~), acetate (CH3CO2"), trifluoroacetate (CF3CO2"), and trifluoromethanesulphonate (CF3SO3 “), perchlorate (CIO4'), a nitrate ion (NO3"), a nitrite ion (NC>2~), phosphate, tetraphenylate, trisphate, tosylate and/or methylsufonate (CH3SC>3‘), preferably chloride or hexafluorophosphate. More preferred as chloride and hexafluorophosphate anions.
The present invention preferably relates to a compound of the general formula:
More preferably yet, the present invention relates to a compound is selected from:
More preferably, the compound is a complex based on a tetradentate ligand H2bapbpy (6,6'-bis[N-(pyndyl)-l-amino]-2,2'-bipyridine) coordinated to Pd (II) or Pt (II).
Applicants found that while palladium complexes as analogues of platinum complexes are typically disregarded as anticancer drugs due to their unfavorably fast ligand exchange kinetics, which may be a relevant feature crucial for its success as a homogenous catalysts for many coupling reactions. However, [Pd(H2bapbpy)](PF6)2 ([3](PF6)2), and [Pt(H2bapbpy)](PF6)2 (|4](PF6)2) were found to exhibit strikingly different biological activities: whereas [2]CI2 harbours a moderate biologically activity, [4](PF6)2 exhibits high cytotoxicity comparable to cisplatin, while [3](PF6)2 shows activities against a wide range of human cancer cell lines in the nanomolar range. Remarkably, the palladium compound [3](PF6)2 was not only found to be much more cytotoxic than its platinum analogue
[4](PF6)2; it appeared to exert a different mechanism of action. Accordingly, it represents the first palladium anticancer drug that causes a complete collapse of the cell redox balance and shows promising in vivo anticancer activity.
It is believed that the biochemical effects of the subject metallodrugs such as those disclosed herein are not limited to DNA-damage induced apoptosis, but may include coordination of the biologically available thiol glutathione to e.g. the cisplatin centre may be associated with detoxification. Cisplatin can also disturb cellular homeostasis and cell redox balance, and increases the presence of reactive oxygen species (ROS). In this context, the present metallodrugs are specifically designed to disturb the intracellular redox balance have been developed as anticancer compounds. As cancer cells already exhibit high metabolic activity and low level of antioxidants they have an increased sensitivity toward such perturbations.
The present invention also relates to a composition comprising a compound as defined herein above. Preferably, the composition further comprises a suitable solvent.
Preferably, the solvent comprises acetonitrile, water, methanol, ethanol or combinations thereof.
The present invention also relates to a pharmaceutical composition comprising a compound, and a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier.
In a further aspect, the present invention also relates to a compound for use as a cytotoxic agent and/or for use as an anti-cancer agent, preferably for cisplatin-resistant cancer cells.
In a further aspect, the present invention also relates to a composition for use in therapy.
In a further aspect, the present invention also relates to a compound, or a composition according to the invention, for use in treating cancer or a high growth infection, preferably testicular cancer, bladder cancer, lung cancer, colon cancer, skin cancer or ovarian cancer in a subject in need thereof.
In a further aspect, the present invention also relates to a method of treating a disease, wherein the method comprises administering a therapeutically effective amount of a compound as defined herein above, or a composition as defined herein, to a subject in need thereof. Preferably, the disease comprises testicular cancer, bladder cancer or ovarian cancer.
In a further aspect, the present invention also relates to the use of a compound or a composition comprising the compound according to the invention, for the manufacture of a medicament for the treatment of a disease in a subject in need thereof. Preferably, the disease is selected from at least one of: testicular cancer, bladder cancer or ovarian cancer, in particular in a platinum resistant cancer.
The choice of the metal centre may have a strong influence on the biological activity of the complex in vitro and in vivo, on its cellular uptake, and on its pKa.
Accordingly, in a further aspect, the present invention also relates to the in vitro, or in vivo use of a use of a compound or a composition comprising the compound according to the invention, for determining if the compound or composition permits retarding or stopping the growth of uncontrolled growth cells, and for retarding or stopping the growth of uncontrolled growth cells.
In a further aspect, the present invention also relates to a method of making a compound according to the invention, comprising the step of combining: i) a ligand of Formula II, or a derivative thereof; with ii) a metal salt selected from palladium, platinum, and/or gold, wherein ligand i) and metal salt ii) form the compound; and optionally isolating, and optionally formulating the isolated or obtained compound into a medication. Preferably, the metal is selected from palladium, platinum, or gold.
The following, non-limiting examples will illustrate the invention further.
General procedures
Chemicals were bought from Alfa Aesar (I^PtCU), Acros organics (^PdCU), and Sigma-Aldrich. Reactions were performed under a nitrogen atmosphere using standard Schlenk techniques. NMR spectra were recorded on a Bruker DPX-500 spectrometer. Chemical shifts are indicated in ppm relative to TMS. Mass spectra were recorded by using a Thermoquest Finnagen AQA Spectrometer. UV-vis experiments were performed on a Cary Varian spectrometer. pH measurements were performed using a PHM220 lab pH meter. Metal concentrations for logP determination were measured on a Vista MPX ICP-OES.
Example 1: Synthesis of [11CI? (formula (V)):
In a 2-necked round bottom flask K2PdCU (304 mg, 0.72 mmol) and Hbbpya (171 mg, 0.53 mmol) were added into a degassed ethanol-water mixture (7:3,100 mL) preheated at 80 °C. The resulting yellow suspension was stirred overnight at 80 °C in a nitrogen atmosphere resulting in a dark green/black suspension. The solution was cooled down to room temperature, filtered, and from the filtrate all solvents were rotary evaporated. The solids were dissolved in MeOH (5 ml) and purified by size exclusion chromatography (Sephadex L-20) using MeOH as eluent. The yellow band was collected and MeOH was removed by rotary evaporation, which resulted in [1]CI?. as a yellow powder. Yield: 42% (111 mg). 2W NMR (500 MHz, DiO, 293 K, in ppm): δ = 8.55 (d, J = 5.5 Hz, 2H, H5'), 8.36 (m, 2H, H3',H6'), 8.19 (t, 3 = 8.0 Hz, 2H, H4), 8.00 (d, i = 7.5 Hz, 2H, H3), 7.71 (m, 2H, H4'), 7.44 (d, J = 8.5 Hz, 2H, H5}.13 C NMR (126 MHz, D20, 293 K, in ppm): δ = 155.5 (C6), 152.0 (C2), 148.7 (Cy), 145.1 (C2), 143.0 (C3'), 142.4 (C4), 128.4 (C4'), 124.6 (C6*), 118.3 (C3), 118.2 (C5). High resolution ES MS m/z (calc): 430.0286 (430.0284 = [Μ ~ H - 2xCI)+). Elem. Anal. Calcd. For CMHisNsPd + MeOH + H20: C, 45.63; H, 3.83; N, 12.67. Found: C, 45.11; H, 3.94; N, 12.86.
Example 2: Synthesis of [2]Cb (formula (VI)):
In a 2-necked round bottom flask K2PtCU (300 mg, 0.730 mmol) and Hbbpya (200 mg, 0.590 mmol) were added into a degassed ethanol-water mixture (3:2,100 mL) preheated at 80 °C. The resulting yellow suspension was stirred overnight at 80 °C in a nitrogen atmosphere resulting in a dark red suspension. The suspension was cooled to room temperature, subsequently cooled on ice, and then filtered. From the filtrate all solvents were rotary evaporated. The solids were dissolved in MeOH (5 mL) and purified by size exclusion chromatography (Sephadex L-20) using MêOH as eluent. The yellow band was collected and MeOH was removed by rotary evaporation, which resulted in [2]Ch as a a red/orange powder. Yield 42% (155 mg), aW NMR (500 MHz, CDaQD and DCI, 293 K, in ppm): 6 = 8.99 (d, J = 5.5 Hz, 2H, H6'), 8.58 (d,J = 7.5 Hz, 2H, H3 ), 8.50 (t, 1 = 7.5 Hz, 2H, H4), 8.39 (t, 1 = 8.0 Hz, 2H, H4), 8.23 (d, J = 6.5 Hz, 2H, H4'), 7.99 (m, 2H, H5'), 7.77 (d, J = 8.5 Hz, 2H, H5). 13C NMR (126 MHz, 293 K, CD3OD and DCI): 6 = 156.4 (C6), 152.4 (C2), 150.4 (C5 ), 144.4 (C2'), 143.7 (C3 ), 142.4 (C4), 129.6 (C4 ), 125.6 (C5'), 119.9 (C3), 118.8 (C3) ppm. High resolution ES MS (H20) m/z (calc.): 519.0893 (519.0897 = [M - H - 2xCI]+). Elem. Anal. Calcd. ForC2oCIHMN5Pt + Yz H20: C, 41.93; H, 2.82; N, 12.22. Found: C, 41.78; H, 3.12; N, 13.51.
Example 3: Synthesis of [31(PF6)2 (See Figure 4):
In a 2-necked round-bottom flask fofPdCU] (80 mg, 0.24 mmol), H2bapbpy (200 mg, 0.58 mmol,obtained as disclosed in S. Bonnet, M. A. Siegler, J. Sanchez Costa, G. Molnar, A. Bousseksou, A. L. Spek, P. Gamez and J. Reedijk, Chem. Commun., 2008, 5619-5621) and degassed ethanol-water mixture (3 :2; 30 mL) were added. The brown suspension was stirred for 2 hours at 80 9C under nitrogen, and an additional fafPdCU] (80 mg, 0.24 mmol) was added. Again, the brown suspension was stirred for 2 hours at 80 eC under nitrogen, and an additional fofPdCU] (80 mg, 0.24 mmol) was added. After full conversion of hhbapbpy was confirmed by TLC (eluens = Acetone/H20/KNC>3 (sat.), (100:10:1)) a precipitate was filtered off and by addition of 1 mL HCI (1.0 M), it was made sure that the solution was acidic. Then, a saturated aqueous solution of KPF6 (5 mL) was added to the filtrate inducing the precipitation of a yellow solid. The suspension was filtered over a membrane filter and the complex [Pd(H2bapbpy)](PF6)2 was obtained as a yellow powder and dried in vacuo.
Example 4: Synthesis of [31(0)2:
To obtain the chloride salt [Pd(H2bapbpy)](PFs)2 (50 mg) was dissolved in acetonitrile (250 mL) and heated a clear solution was obtained. Then, concentrated HCI (37% w/v, 1 mL) was added upon which a precipitate appeared. The suspension was filtered over a membrane filter and the complex [Pd(H2bapbpy)]CI2 was obtained as a yellow powder and dried in vacuo.
Example 5: Synthesis of [4](PF6)2:
In a 2-necked round-bottom flask K2[PtCI4] (101 mg, 0.24 mmol), H2bapbpy (200 mg, 0.58 mmol) and degassed ethanol-water mixture (7 : 3; 30 mL) were added. The brown suspension was stirred for 2 hours at 80 sc under nitrogen, and an additional K2[PtCU] (101 mg, 0.24 mmol) was added. Again, the brown suspension was stirred for 2 hours at 80 ?C under nitrogen, and an additional K2[PtCI4] (101 mg, 0.24 mmol) was added. After full conversion of H2bapbpy was confirmed by TLC (eluens = Acetone/H20/KN03 (sat.), (100:10:1)) a precipitate was filtered off and an aliquot of HCI (1.0 Μ, 1 mL) was added to make the solution acidic. Then, a saturated aqueous solution of KPFe (5 mL) was added to the filtrate inducing the precipitation of a yellow solid. The suspension was filtered over a membrane filter and the complex was obtained as a yellow powder and dried in vacuo. The product obtained here is [Pt(H2bapbpy)](PF6)2 Example 6: Synthesis of [41(0)2:
To obtain the chloride salt (Pt(H2bapbpy))(PF42 was dissolved in acetonitrile and heated to ensure a better and full solubility - typically 50 mg of complex in 250 mL of acetonitrile. Then, to keep concentrations of water low, 1 mL of HCI (37% w/v) was added upon which the product precipitated. The suspension was filtered over a membrane filter and the complex [Pt(H2bapbpy)]C!2 was obtained as a yellow powder and dried in vacuo.
Example 7: pKa determination pH titration: 6 mL of a 67 μΜ solution of [1]CI2 or [2]CI2 in hydrochloric acid (0.033 M) was put in a 15 mL vial. A pH measurement electrode was added and when stable the pH was logged, aliquots of aqueous NaOH (0.05 - 5M) were added to give a range of pH values while stirring. After each stable pH a UV-vis absorbance spectrum was recorded. The relative concentration of [MfHbbpya]2* was calculated using the Lambert-Beer law and then plotted vs. pH. The pKa was determined by modelling the curve using a simplified two-parameter Hill-
slope equation:
.Log P value determination: 1.00 mM stock solutions of [1]CI2 and [2]CI2 in octanol-saturated water were prepared. 0.2 mL aliquots were transferred to 15 mL corning centrifugation tubes and diluted with octanol-saturated water to 1.0 mL, to give 0.2 mM solutions (in threefold). Then, 1.0 mL water-saturated octanol was added to each solution (not for the control samples, which remained at a volume of 1 mL). The solutions were shaken for 60 minutes on a GFL 3016 shaker at maximum speed and centrifuged for 10 minutes at RCF = 2800 g at T = 293 K. Then, 100 pL of the aqueous phase (in threefold) was transferred into a vial, and 1.0 ml 65% nitric acid was added to degrade the compounds overnight. 0.8 mL of the resulting solution was then diluted with 11 mL MilliQ water in corning tubes. Then the metal concentration of each sample was measured using ICP-JOS, and the initial equilibrium metal concentration in the water and octanol phases was calculated from the dilution factors. Finally, the logP value was calculated using the following equation:
Example 8: Cell culturing and ECso cytotoxicity assay
Cellular uptake of [1]CI2, [2]Cb, [3](PF6)2, and [4](PF6)2
Uptake studies for complexes [1]CI2, [2]CI2, [Pd(H2bapbpy)](PFe)2 ([3](PFe)2) and[Pt(H2bapbpy)](PF6)2 ([4](PF6)2) were conducted on A375 and A549 lung cancer cells. 10 χ 103 A375 cells and 17 χ 103 A549 cells were seeded at t = 0 h in Opti-MEM complete in each well of a 6-well plate. At t = 48 h cells were treated with complexes to give a final concentration close to the ECso values (1.9,1.7, 0.05 or 0.2, and 1.0 μΜ for respectively [1]CI2, [2]CI2, [3](PF6)2, and [4](PF6>2) in the dark after 24 h in a total volume of 4 mL. After 24 h of drug incubation at 37 °C, the medium was aspirated, the cells were washed with PBS-buffer, trypsinized, counted using BioRad Cell Counting Slides on a BioRad TC10 automated cell counter, and pelleted by centrifugation (700 χ g, 5 min). The supernatant was removed, and each sample was digested overnight in concentrated nitric acid at room temperature (> 65%). MilliQ water was added to each sample to obtain a final concentration of 5% HNO3. For ICP-MS measurements, the system was optimized with a palladium-platinum solution. The calibration range was from 0 to 25 pg/l, and obtained detection limit for all isotopes was 0.01 pg/l. Silver and indium were used for internal standard, to correct for sample-dependent
matrix effects. No reference sample was available; therefore several samples were spiked with a known concentration. The recoveries of the spiked concentrations were all within a 10% deviation.
Example 9: Synthesis and crystal structure
In a preferred embodiment of the subject invention, the square-planar complexes [Pd(Hbbpya)]Ö2 ([l]Ch) and [Pt(Hbbpya)]Cl2 ([2]Ch) were synthesized by reacting ligand Hbbpya with fofPdCU] and falPtCU], respectively, in an ethanol-water mixture (Figure 3).
After purification using size exclusion chromatography, analysis with NMR spectroscopy, elemental analysis, and high resolution mass spectrometry confirmed that the compounds were analytically pure. Vapour diffusion of acetone into a solution of [l]Ch in methanol yielded yellow-colored crystals suitable for X-ray diffraction studies (Figure 3, schematic synthesis of [l]Ch and [2]Cb, and Figure 6, X-ray crystal structure of (l]Ch)
The compound [l]Ch crystallizes with one molecule of MeOH. Pdl-Nl and Pdl-N5 bond distances of the terminal pyridyl groups are 2.0600(18) and 2.0680(18) Λ, respectively (Error! Reference source not found.), whereas the Pdl-N2 and Pdl-N4 bond distances of the internal pyridines are 1.9887(10) and 1.9933(18) A, respectively, thus much shorter. These trends are similar to those in [Pd(H2bapbpy)](PF5)2 (PKPFeh), but are even more pronounced in the analogue palladium(ll) complex without any amine bridge, i.e. [Pd(qtpy)](PF6)2.
Table 4: Selected bond lengths (A) in the crystal structures of [1]CI2, of the H2bapbpy analogue [3](PF6)2, and of [Pd(qtpy)](PF6)2.
b: Values taken from Constable et al.12 c: Average value of the four coordinating Pd-N bonds in each complex.
The differences between the open and closed side of the tetrapyridyl ligands are also apparent when comparing the bond angles. For [ljCb the Nl-Pdl-N5 angle on the open side is 108.84(7)°, while the angle N2-Pdl-N4 on the closed side is 93.22(7)°, thus more than 15° smaller. In [3](PF6>2 and [Pd(qtpy)](PFe)2 bond internal angles N3-Pdl-N4 and Nl-Pdl-N5 are 82.19(7) and 81.2°, respectively. These differences can be explained by the formation of one 6-membered ring and two 5-membered rings with Flbbpya, while coordination of Fhbapbpy generates one 5-membered ring and two 6-membered rings, and qtpy generates only 5-membered rings. Such differences increase the opening of the ligands as follows: Fhbapbpy < Hbbpya < qtpy: in [3](PF6)2 N6-Pdl-Nl is 96.38(7)°, in [1]CI2 Nl-Pdl-N5 is 105.84(7)°, and in [Pd(qtpy)](PF6)2 N2-Pdl-N2B is 117.8°.
Table 5: Selected bond angles, torsion angles (°), and τ4 values for [1)02, [3](PF6)2, and [Pd(qtpy)](PF6)2.
* T4 is obtained using the equation
; a value of 0 corresponds to a square- planar coordination geometry and a value of 1 to a tetrahedral coordination geometry, a Values taken from Chapter 6 b Values taken from Constable et al.12
As a result of the different chelating rings generated by the three ligands H2bapbpy, Hbbpya, and qtpy, the distortions of the square-planar coordination sphere of the metal also greatly vary. A perfect square-planar coordination geometry can be defined as a four-coordinate complex with ligands that coordinate in a single plane, and it should contain bond angles of only 90 and 180°. The X4 value introduced by Houser and co-workers
quantifies the square-planarity of tetracoordinated metal complexes by distinguishing complexes with a u value of 0 that are perfectly square-planar, and complexes with a 14 value of 1 that have a perfect tetrahedral geometry.
Using the crystal structures of [ljCb, [3](PFe)2, and [Pd(qtpy)](PF6h t4 values of respectively 0.090, 0.193, and 0.260 (Table 5) were calculated indicating that [l]Ch is by far the most square-planar geometry, while [3](PF6)2 and [Pd(qtpy)](PF6)2 have both severely distorted geometries. The torsion angle of the coordinating nitrogen atoms is a measure of the degree of flatness of the coordination plane of square-planar complexes. For [l]Ch, [3](PF6)2, and [Pd(qtpy)](PF6h these torsion angles are 0.20,17.30, and 0.54°, respectively. Thus, whereas in [1]2+ and [Pd(qtpy)]2+ the tetrapyridyl ligands are coordinated almost in one single plane, in [3]2+ the ligand assumes a more helical geometry. To conclude, [l]Ch is highly square-planar and very flat. The distance between the hydrogen atoms of Cl and C20 is 1.905 A, which is apparently long enough to prevent distortion of the coordinated Hbbpya ligand.
Previously, Hbbpya has already demonstrated a high degree of planarity in octahedral iron(ll) complexes, and preliminary data shows that it is also highly planar in nickel(ll) and ruthenium(ll) complexes.
This planar geometry significantly differs from the H2bapbpy ligand that coordinates to metal ions in a distorted fashion. Overall, the removal of an amine bridge in H2bapbpy, results in the ligand Hbbpya that can coordinate to palladium(ll) or platinum(ll) with a highly square-planar geometry.
Compounds [l]Cb and [2]Ch were found to have mildly to strong cytotoxic properties. Without wishing to be bound to any particular theory, it was originally believed that the similar coordination mode of Hbbpya and H2bapbpy to palladium(ll) and platinum(ll) ions may permit to predict similar activities for these metal complexes in vitro.
To check this hypothesis, also the cytotoxicity of the several Hbbpya and H2bapbpy metal complexes were tested in vitro against three different cancer cell lines (A375 skin, A549 lung, and MDA-MB231 breast cancer) and one non-malignant lung cell line (MRC-5). The cell-testing protocol was adapted from S. L. Hopkins, B. Siewert, S. H. C. Askes, P. Veldhuizen, R. Zwier, M. Heger and S. Bonnet, Photochem. Photobiol. Sci., 2016,15, 644-653. 24 h after seeding, the cells were incubated for 24 h with the drug, the media was refreshed, and the cells were further incubated with drug-free medium for 48 h. A sulfo-rhodamine B (SRB) cell-counting assay was performed at 96 h, and effective concentrations (EC50) were determined by comparing cell viability of treated wells with drug-free control wells. The EC50 values are summarized in Table 6. EC50 values for A375 and A549 cancer cell lines for [ljCh and [2]Ch were similar and ranged between 1.6-1.9 μΜ. For MDA-MB231 and MRC-5 cancer cells both compounds showed lower cytotoxicty in the 4.7-6.8 μΜ range. Overall, the cytotoxicity of [l]Ch and [2]Ch was found comparable with that of cisplatin, i.e. in the low micromolar range. The cytotoxity of [2]Ch is comparable with that of [Pt(H2bapbpy)](PF6)2 ([4](PF6)2, but the cytotoxicity of [l]Ch was nowhere near the exceptional activity of [3](PFe)2, that showed EC50 values below 0.2 μΜ for all the cell lines tested in this panel. Although the ligands are highly similar, once coordinated to palladium they have highly different biological properties as their palladium(ll) complexes.
Table 6: Cytotoxicity expressed as cell growing inhibition effective concentrations (EC50 with 95% confidence intervals, in μΜ) for [1]CI2, [2]CI2, and cisplatin on skin (A375), lung (A549), and breast (MDA-MB231) cancer cell lines, and on non-cancerous lung fetal cell line (MRC-5).Cell Line
The cytotoxicity of the two complexes [3](PF6)2 and [4](PF6)2 was tested against five different cancer cell lines (A375, A431, A549, MCF-7, MDA-MB231} and one non-malignant lung cell line (MRC-5).
The cell testing protocol was adapted from Hopkins et al. as set out above. The cytotoxicity results for compounds [3](PF6)2 and [4);(.PF6)2 are summarized in Table 7.
Table 7: Cytotoxicity expressed as cell growing inhibition effective concentrations (EC50 with 95% confidence intervals, in μΜ)
For [4](PF6)2 EC50 values between 0.80 μΜ for MCF-7 cells and 1.4 μΜ for MRC-5 cells were found, which makes the platinum complex [4](PF6)2 slightly more cytotoxic than cisplatin, For |3](PF6)2 the EC50 values ranged between 0.058 μΜ for A375 cells and 0.19 μΜ for A549 cells, which is remarkably low. This makes the compound [3](PF6)2 one of the most cytotoxic palladium compounds known to date, as it is 520 Times more toxic than its platinum analogue [4](PF6)2 or cisplatin over the full panel of tested cell lines in spite of the almost identical structural features.
Example 10: Cellular Uptake
Considering the different in vitro activity of [l]Cb and [4](PF6)2 uptake studies were performed to determine whether the differences in cytotoxicity are caused by different
intracellular concentrations of both palladium compounds, A375 or A549 cells were seeded, and treated with (l]Cb, [2]Ö2, [SKPFeh, [4]{PF6}2^ or cisplatin at their ECso concentrations. After 24 h drug intubation, cells were washed, trypsinized, counted, digested and the amount of metal content was measured using ICP-MS (see Experimental section for the full procedure). [l]Cb and {2)CI2 showed similar uptake by A375 or A549 cells (7.2 and 9.4 nmol/106 cells) with an uptake efficiency of 8 and 16%. [3}(PF6)2 showed an uptake of 1.2 (54%) and 5.3 nmol Pd/106 cells (39%) for A375 and A549 cells, respectively. For [4]{PF&)2a similar trend was observed with 19 and 22 nmol Pt/106 cells for A375 and A549 cells, respectively, corresponding to an uptake efficiency of 58% in both cell lines. Cisplatin, by comparison, showed very low metal uptake (0.06 and 0,07 nmol Pt/106 cells for A375 and A549 cells, respectively), which demonstrates that once in the cell cisplatin is much more harmful than the tetrapyridyl complexes. The lower cytotoxicity of the Hbbpya complexes compared to the Fhbapbpy complexes does not Seem to be related to a lower metal uptake: treatment at the ECso the uptake values were overall similar. Compound [4](PFö)2 may owe its higher cytotoxicity compared to [l)Ch and [2]Ch due to a higher uptake, but [3](PFs)2 clearly has a much lower metal uptake and much higher cytotoxicity. These uptake results emphasize the remarkable cytotoxicity of [3](PFe)2 as its cellular concentrations in this experiment are the lowest by far, while it exhibited the highest cytotoxicity.
Table 8: Cellular uptake for A375 and A549 for compounds [1]CI2, [2]CI2, [3](PF6)2, [4]{PF6)2, and cisplatin.
Example 11; Lipophilicity and pKa in medicinal chemistry higher uptake and improved cytotoxicity are often associated with higher lipophilicitiy. The octanoi-water partition coefficient (logP value) is widely used as an indicator of lipophilicity (positive logP value) or hydrophiilelty (negative logP value).
The PFg salts of the Fbbapbpy complexes were first converted to [3)02 and [4)02, respectively, as DMSO would be required for initial solubilization of the PFe salts.
The logP values for complexes [l]Cb to [4]Cb are shown in
Table. The logP value for [4]Ch (-0.45) was the highest of all the complexes, and also corresponded with the highest cellular uptake of [4](PFg)2- [3)02 with a logP value of -1.63 is the most hydrophilic of the compounds, and as the PFe salt was taken up second highest. The logP values of [l]Cb and [2]Cb were found to be -0.96 and -1.59, respectively. In conclusion, all four tested complexes are rather hydrophilic, and no obvious relationship between metal uptake and logP could be observed for this series of complexes.
Each amine bridge in the tetrapyridyl ligands Fbbapbpy and Hbbpya bears an acidic proton. Consecutive deprotonation of the two amine bridges in Fbbapbpy can result in dicationic, monocationic, or neutral complexes, whereas the single amine bridge in Hbbpya complexes can only lead to dicationic or monocationic species. Of course, the acid-base equilibrium may influence both the logP values and the uptake of these complexes in vitro. The pKa for the first deprotonation of [3]2+ and [4}2+ is 7.8 and 8.3 in water, respectively.
For [1]2+ and [2]2+the pKa in water was also determined by monitoring with UV-vis Spectroscopy the titration of [l]Cb and [2]Cb in HCI (33 mM) with NaOH. Under acidic conditions the UV-vis spectra of [1)2+ and [2]2+ only show absorption in the visible region with a tail up to 423 nm and 425 nm, respectively (green curves in Figure 6). Upon base addition a significant increase in absorbance was observed, characterized by a new absorption maximum at 416 nm for [1]2+ and 430 nm for |2p; Simultaneously, a major decrease in absorbance was found at 261 and 362 nm for [1)2+ and at 239 and 372 nm for [2]2+. The evolution of the absorbance spectra for both complexes occurred with isosbestic
points at 316 and 397 nm for [1]2+, and at 312 and 405 nm for [2]2+, indicating that only one reaction takes place. Further basification did not lead to any further change in the absorbance. The pH-dependence of the absorbance spectra of [1]2+ and [2]2+ in water can be explained by the deprotonation of [1]2+ or [2]2+ to form [Pd(bbpya)]+ or [Pt(bbpya)]+, respectively. The corresponding pKa was determined to be 5.5 and 4.6 for [1]2+ and [2]2+, respectively. Thus, [1]2+ and [2]2+ have much lower pKa's compared to [3]2+ and [4]2+. Such higher acidity can be explained by a difference in steric repulsion: Hbbpya complexes have an almost perfect square-planar geometry (τ = 0.090), whereas complexes of hhbapbpy are much more distorted (τ = 0.193, Figure 6). As a result the negative charge located on the deprotonated bbpya" ligand in [Pd(bbpya)]+ and [Pt(bbpya)]+ is much better delocalized than the negative charge located on Hbapbpy" in [Pd(Hbapbpy)]+ and [Pt(Hbapbpy)]+.
Another reason for the lower pKa of [1]2+ and [2]2+ compared to [3]2+ and [4]2+ may be that the negative charge of the [Pd(bbpya)]+ and [Pt(bbpya)]+ complexes may be delocalized over two bpy moieties, whereas that of the [Pd(Hbapbpy)]+and [Pt(Hbapbpy)]+ complexes may be delocalized over only one bpy moiety and two pyridyl fragments.
Overall, at physiological pH (7.4) the Hbbpya complexes will be fully deprotonated, thus monocationic, whereas the Hzbapbpy complexes will be mixtures of dicationic and monocationic species (see Table 9).
Table 9: LogP and pKa values for [1]CI2, [2]CI2, [3]CI2, and [4]CI2.
Although the tetrapyridyl ligands Hbbpya and Hzbapbpy structurally are highly similar, the coordination to palladium(ll) or platinum(ll) centers results in complexes with significant differences in coordination geometry, acidity, cellular uptake, and cytotoxicity.
The crystal structures of the palladium(ll) complexes with qtpy, Hbbpya, or hhbapbpy show that the palladium Hbbpya complex is the most square-planar as well as the flattest of the three complexes. In the H2bapbpy complexes electronic conjugation in the distorted ligand is less possible, making deprotonation less favorable compared to Hbbpya complexes.
As a result [1]2+ and [2]2+ will not be present as dicationic species in vitro whereas [3]2+ and [4]2+ are present in both dicationic and monocationic forms.
Without wishing to be bound to any particular theory, it is hypothesized that the complexes described in this chapter are taken up via passive diffusion, both the charge state and the lipophilicity may play a key role.
Specifically in case of the H2bapbpy complexes, their pKa values close to physiological conditions makes the complexes flexible regarding charge-dependent biochemical interactions with for instance negatively charged DNA.
While observed differences in cytotoxicity may advantageously be explained by differences in uptake for [ljCh, [2]Cl2, and [4](PF6h, provided that these complexes have similar mechanism of actions, it was however found that the cytotoxicity and uptake of [3] (PFe)2 demonstrates that it is orders of magnitude more cytotoxic, and thus efficient in killing the cell once taken up, and that it may have a distinct mechanism of actions compared to the other complexes.
[l]Ch and [2]Ch, albeit less cytotoxic than [3]{PFe)2, also show that the class of non-cyclic tetrapyridyl complexes is promising as anticancer agents when the ligand is functionalized with a single non-coordinating amine bridge. These complexes also advantageously illustrate that non-coordinated chloride counter-anions, combined with the amine bridges, may result in much more water-soluble complexes, as compared to cisplatin.
Example 12: In vivo experiments
The in vivo anticancer activity of [3](PF6)2 and [4](PF6)2 was evaluated in CT-26 colon cancer-bearing mice as disclosed in J. Mayr, P. Heffeter, D. Groza, L. Galvez, G. Koellensperger, A. Roller, B. Alte, M. Haider, W. Berger, C. R. Kowol and B. K. Keppler, Chem. Sci., 2017, 8, 2241-2250.
Treatment existed of equimolar dosages [3](PF6)2 (2.5 mg/kg) or [4] (PF6)2 (2.8 mg/kg), which was found to be the maximum-tolerated dose for [3](PF6)2. The complexes were administered by intraperitoneal injections for 5 times (on day 3, 5,7,10, and 12) and mice were sacrificed 24 h after the last application.
The impact of the metal complexes on tumour growth was determined by measuring the tumour volume (mm3) and the results are shown in Figure 6.4. Compared to untreated control BALB/c mice, [4](PF6)2 treatment had no effect on tumour growth, indicating that in this treatment regime [4](PF6)2 is inactive at the doses used. In contrast, [3](PF6)2 induced from the first day a significant delay in tumour growth and on day 12 a 37% reduction compared to the control group. Thus, as previously witnessed in in vitro experiments, treatment with [3](PF6)2 or [4](PF6)2 leads to different activity, despite structural similarities, also under in vivo conditions. H2bapbpy complexes [3](PF6)2, and [4](PF6)2 demonstrate very diverse cytotoxicity patterns. Exposure of various cancer cell lines to the drugs results in ECso values for [3](PF6)2 in the nanomolar range, and for [4](PF6)2 in the submicromolar to low micromolar range.
Example 13: synthesis of fAu(bapbpy)lPFe h^bapbpy (34 mg, 0.10 mmol) was first dissolved in hot ethanol (30 mL) and filtered to make a clear solution of Fbbapbpy. In a 2-necked round-bottom flask NaAuCU (61 mg, 0.15 mmol), excess KPFe (2 mmol, 368 mg) and the ethanol solution of Fhbapbpy were added. The initially yellow solution was stirred for 4 hours at 60 eC under nitrogen, upon which it turned into a red suspension. The suspension was filtered over a membrane filter and the solid washed by water and ether, then dried in vacuum. The complex [Au(bapbpy)](PF6) was obtained as a red powder in 38% yield (26 mg). 1H NMR (400 MHz, [D6JDMSO) δ: 8.01 (2 H, dd, J = 8.6, 7.3), 7.86 (6 H, dddd, J = 8.0, 7.0, 5.6, 1.5), 7.37 (4 H, td, J = 8.5,1.5), 6.93 (2 H, td, J = 6.7,1.7). MS: 535.6 [Au(bapbpy)]+.
Claims (26)
Priority Applications (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| NL2019752A NL2019752B1 (en) | 2017-10-18 | 2017-10-18 | Cytotoxic Complexes |
| PCT/NL2018/050682 WO2019078715A1 (en) | 2017-10-18 | 2018-10-18 | Cytotoxic complexes |
Applications Claiming Priority (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| NL2019752A NL2019752B1 (en) | 2017-10-18 | 2017-10-18 | Cytotoxic Complexes |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| NL2019752B1 true NL2019752B1 (en) | 2019-04-25 |
Family
ID=61224463
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| NL2019752A NL2019752B1 (en) | 2017-10-18 | 2017-10-18 | Cytotoxic Complexes |
Country Status (2)
| Country | Link |
|---|---|
| NL (1) | NL2019752B1 (en) |
| WO (1) | WO2019078715A1 (en) |
Families Citing this family (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2019084696A1 (en) * | 2017-11-06 | 2019-05-09 | Queen's University At Kingston | Platinum compounds for binding guanine quadraplexes |
| US11932660B2 (en) | 2020-01-29 | 2024-03-19 | Universal Display Corporation | Organic electroluminescent materials and devices |
Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN107163087A (en) * | 2017-07-13 | 2017-09-15 | 长春海谱润斯科技有限公司 | A kind of organometallic complex, synthetic method and its organic luminescent device |
-
2017
- 2017-10-18 NL NL2019752A patent/NL2019752B1/en not_active IP Right Cessation
-
2018
- 2018-10-18 WO PCT/NL2018/050682 patent/WO2019078715A1/en not_active Ceased
Patent Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN107163087A (en) * | 2017-07-13 | 2017-09-15 | 长春海谱润斯科技有限公司 | A kind of organometallic complex, synthetic method and its organic luminescent device |
Non-Patent Citations (6)
| Title |
|---|
| CHANGCHUN HAIPU RUNSI TECH CO LTD: "Display of compounds in CN107163087", CN107163087, 15 September 2017 (2017-09-15), pages 1 - 19, XP055473328 * |
| HE LEI ET AL: "Aza-bridged bisphenanthrolinyl Pt(II) complexes: Efficient stabilization and topological selectivity on telomeric G-quadruplexes", JOURNAL OF INORGANIC BIOCHEMISTRY, ELSEVIER INC, US, vol. 166, 9 November 2016 (2016-11-09), pages 135 - 140, XP029845411, ISSN: 0162-0134, DOI: 10.1016/J.JINORGBIO.2016.11.011 * |
| THOMAS BARK ET AL: "[1,10]-Phenanthrolin-2-yl Ketones and Their Coordination Chemistry", INORGANIC CHEMISTRY, vol. 44, no. 24, 1 November 2005 (2005-11-01), EASTON, US, pages 8733 - 8739, XP055473342, ISSN: 0020-1669, DOI: 10.1021/ic050996k * |
| V. H. S. VAN RIXEL ET AL: "Green light-induced apoptosis in cancer cells by a tetrapyridyl ruthenium prodrug offering two trans coordination sites", CHEMICAL SCIENCE, vol. 7, no. 8, 1 January 2016 (2016-01-01), United Kingdom, pages 4922 - 4929, XP055473312, ISSN: 2041-6520, DOI: 10.1039/C6SC00167J * |
| VINCENT H. S. VAN RIXEL ET AL: "Controlling with light the interaction between trans -tetrapyridyl ruthenium complexes and an oligonucleotide", DALTON TRANSACTIONS, vol. 47, no. 2, 23 November 2017 (2017-11-23), GB, pages 507 - 516, XP055473314, ISSN: 1477-9226, DOI: 10.1039/C7DT03613B * |
| ZULEMA ARCIS-CASTÍLLO ET AL: "Tuning the Transition Temperature and Cooperativity of bapbpy-Based Mononuclear Spin-Crossover Compounds: Interplay between Molecular and Crystal Engineering", CHEMISTRY - A EUROPEAN JOURNAL, vol. 17, no. 52, 23 December 2011 (2011-12-23), DE, pages 14826 - 14836, XP055474212, ISSN: 0947-6539, DOI: 10.1002/chem.201101301 * |
Also Published As
| Publication number | Publication date |
|---|---|
| WO2019078715A1 (en) | 2019-04-25 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Biancalana et al. | α-Diimines as versatile, derivatizable ligands in ruthenium (II) p-cymene anticancer complexes | |
| Hemmert et al. | Synthesis, structures, and antimalarial activities of some silver (I), gold (I) and gold (III) complexes involving N-heterocyclic carbene ligands | |
| AU2002329097B2 (en) | Inclusion compound comprising cucurbituril derivatives as host molecule and pharmaceutical composition comprising the same | |
| Hotze et al. | Synthesis, characterization, and crystal structure of α-[Ru (azpy) 2 (NO3) 2](azpy= 2-(Phenylazo) pyridine) and the products of its reactions with guanine derivatives | |
| Habtemariam et al. | Control of aminophosphine chelate ring-opening in Pt (II) and Pd (II) complexes: potential dual-mode anticancer agents | |
| Swaminathan et al. | Hinged bipodal furoylthiourea-based Ru (II)-arene complexes: Effect of (ortho, meta, or para)-substitution on coordination and anticancer activity | |
| Muley et al. | Synthesis, structure, spectral, redox properties and anti-cancer activity of Ruthenium (II) Arene complexes with substituted Triazole Ligands | |
| Ermakova et al. | Bioactive mixed-ligand zinc (II) complexes with 1H-tetrazole-5-acetic acid and oligopyridine derivatives | |
| NL2019752B1 (en) | Cytotoxic Complexes | |
| Yusof et al. | Homoleptic tin (IV) compounds containing tridentate ONS dithiocarbazate Schiff bases: Synthesis, X-ray crystallography, DFT and cytotoxicity studies | |
| Kilpin et al. | Organogold (III) complexes containing chelating bis (amidate) ligands: Synthesis, characterisation and biological activity | |
| US20090209046A1 (en) | Neutral Pharmaceuticals | |
| Dömötör et al. | Comparative equilibrium and structural studies of new pentamethylcyclopentadienyl rhodium complexes bearing (O, N) donor bidentate ligands | |
| Alajrawy et al. | In-vitro cytotoxic activity and theoretical investigations for new mononuclear Pt (IV) and dinuclear Ru (III) with o-phenylenediamine ligand complexes against L20B cell line | |
| Büchele et al. | Synthesis, characterization, and biomedical evaluation of ethylene-bridged tetra-NHC Pd (ii), Pt (ii) and Au (iii) complexes, with apoptosis-inducing properties in cisplatin-resistant neuroblastoma cells | |
| US8324197B2 (en) | Transplatinum complexes with N2O2 donor sets as cytotoxic and antitumor agents | |
| Green et al. | Synthetic strategy towards heterodimetallic half-sandwich complexes based on a symmetric ditopic ligand | |
| Asman | Kinetics and mechanistic study of polynuclear platinum (II) polypyridyl complexes; A paradigm shift in search of new anticancer agents | |
| Ratcliff et al. | Part 1: Experimental and theoretical studies of 2-cyano-2-isonitroso-N-piperidynylacetamide (HPiPCO), 2-cyano-2-isonitroso-N-morpholylacetamide (HMCO) and their Pt-and Pd-complexes | |
| Ravera et al. | Synthesis, characterization, structure, molecular modeling studies and biological activity of sterically crowded Pt (II) complexes containing bis (imidazole) ligands | |
| Joshi et al. | A Mo (VI) based coordination polymer as an antiproliferative agent against cancer cells | |
| Calatayud et al. | Synthesis of hybrid ligands derived from benzil, thiosemicarbazide and heteroaromatic hydrazides and their reactivity with group 12 metals | |
| Van Thong et al. | Cyclometalated platinum (II) complexes bearing natural arylolefin and quinolines ligands: Synthesis, characterizations, and in vitro cytotoxicity | |
| EP4069710B1 (en) | Pd(i) complexes, process for their preparation and use thereof as antitumour agents | |
| Shaira et al. | The effect of mixed-metal ruthenium (II)-platinum (II) and cobalt (II)-platinum (II) complexes of tetra-2-pyridyl-1, 4-pyrazine chelate ligands on the substitution of chlorides from platinum metal |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| MM | Lapsed because of non-payment of the annual fee |
Effective date: 20201101 |