The novel steroid conjugates 17 beta-[N-[N'-(2-chloroethyl)-N'-nitroso] carbamoyl]-glycyl-19-nortestosterone (1) and 17 beta-[N-[N'-(2-chloroethyl)-N'-nitroso]carbamoyl]-L-alyanyl-19- nortestosterone (2) were synthesized and characterized with respect to affinity for steroid receptors and for androgenic efficacy. At an i.p. dosage of 50 mg/kg, conjugates 1 and 2 induced strong tumor inhibition of Nobel Nb prostate carcinoma in rats, but also a marked loss of body weight. In two further experiments, treatment with conjugate 2 at a dosage of 25 mg/kg demonstrated high antitumor activity without indication of toxicity. Conjugate 2 achieved the same tumor growth inhibition as a nearly twofold molar dose of cyclophosphamide. The results indicate reproducibly high antitumor activity of 2 in the Noble Nb model at a well tolerated dosage. A low dose equimolar mixture of unlinked N-(2-chloroethyl)-N-nitrosocarbamoyl (CNC)-alanine and 19-nortestosterone was significantly more toxic than conjugate 2, showing about the same adverse effect on the body weight as the conjugate at high dosage. CNC-L-alanine at equimolar dosage was highly toxic, causing early death of all animals.
{"title":"Novel steroid-linked conjugates of 17 beta-[N-[N'-(2-chloroethyl)-N'-nitroso]carbamoyl]amino acids and their antineoplastic activity against Noble Nb prostate carcinoma model in rats.","authors":"W Tang, M R Schneider, G Eisenbrand","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>The novel steroid conjugates 17 beta-[N-[N'-(2-chloroethyl)-N'-nitroso] carbamoyl]-glycyl-19-nortestosterone (1) and 17 beta-[N-[N'-(2-chloroethyl)-N'-nitroso]carbamoyl]-L-alyanyl-19- nortestosterone (2) were synthesized and characterized with respect to affinity for steroid receptors and for androgenic efficacy. At an i.p. dosage of 50 mg/kg, conjugates 1 and 2 induced strong tumor inhibition of Nobel Nb prostate carcinoma in rats, but also a marked loss of body weight. In two further experiments, treatment with conjugate 2 at a dosage of 25 mg/kg demonstrated high antitumor activity without indication of toxicity. Conjugate 2 achieved the same tumor growth inhibition as a nearly twofold molar dose of cyclophosphamide. The results indicate reproducibly high antitumor activity of 2 in the Noble Nb model at a well tolerated dosage. A low dose equimolar mixture of unlinked N-(2-chloroethyl)-N-nitrosocarbamoyl (CNC)-alanine and 19-nortestosterone was significantly more toxic than conjugate 2, showing about the same adverse effect on the body weight as the conjugate at high dosage. CNC-L-alanine at equimolar dosage was highly toxic, causing early death of all animals.</p>","PeriodicalId":7927,"journal":{"name":"Anti-cancer drug design","volume":"13 7","pages":"815-24"},"PeriodicalIF":0.0,"publicationDate":"1998-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"20719557","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The synthesis of four binuclear platinum complexes of general formula ¿cis-[Pt(NH3)2Cl]2(L)¿ (NO3)2 [L is 4,4'-dipyridyl sulfide (compound I), 4,4'-dipyridyl selenide (compound II), bis(3-methyl-4-pyridyl) sulfide (compound III) or bis(3-methyl-4-pyridyl) selenide (compound IV)] has been achieved. These compounds have been characterized by elemental analysis, IR, 1H-NMR, 13C-NMR and 195Pt-NMR spectroscopy. The above dinuclear platinum complexes have been assayed for antitumor activity in vitro against the cisplatin-sensitive L1210 (the mice leukemia) and cisplatin-insensitive HCT8 (the human coloadenocarcinoma) cell lines. The compounds show IC50 values comparable to or higher than cisplatin against L1210 cell line; however, they have lower IC50 values than cisplatin against the cisplatin-insensitive HCT8 cell line. The complexes containing S exhibit a cytotoxicity against the two cancer cell lines superior to the Se analogues. DNA binding studies indicate the compound I possibly interacts with DNA nonintercalatively. The mode of DNA binding of the dinuclear Pt(II) complexes bridged by 4,4'-dipyridyl selenides or sulfides may be different to that of the aliphatic diaminebridged dinuclear Pt(II) complexes reported previously.
{"title":"Synthesis and biological activity of binuclear platinum complexes containing two monofunctional cis-[Pt(NH3)2Cl]+ units bridged by 4,4'-dipyridyl selenides or sulfides.","authors":"G Zhao, H Lin, S Zhu, H Sun, Y Chen","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>The synthesis of four binuclear platinum complexes of general formula ¿cis-[Pt(NH3)2Cl]2(L)¿ (NO3)2 [L is 4,4'-dipyridyl sulfide (compound I), 4,4'-dipyridyl selenide (compound II), bis(3-methyl-4-pyridyl) sulfide (compound III) or bis(3-methyl-4-pyridyl) selenide (compound IV)] has been achieved. These compounds have been characterized by elemental analysis, IR, 1H-NMR, 13C-NMR and 195Pt-NMR spectroscopy. The above dinuclear platinum complexes have been assayed for antitumor activity in vitro against the cisplatin-sensitive L1210 (the mice leukemia) and cisplatin-insensitive HCT8 (the human coloadenocarcinoma) cell lines. The compounds show IC50 values comparable to or higher than cisplatin against L1210 cell line; however, they have lower IC50 values than cisplatin against the cisplatin-insensitive HCT8 cell line. The complexes containing S exhibit a cytotoxicity against the two cancer cell lines superior to the Se analogues. DNA binding studies indicate the compound I possibly interacts with DNA nonintercalatively. The mode of DNA binding of the dinuclear Pt(II) complexes bridged by 4,4'-dipyridyl selenides or sulfides may be different to that of the aliphatic diaminebridged dinuclear Pt(II) complexes reported previously.</p>","PeriodicalId":7927,"journal":{"name":"Anti-cancer drug design","volume":"13 7","pages":"769-77"},"PeriodicalIF":0.0,"publicationDate":"1998-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"20719554","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
N D Henderson, S M Lacy, C C O'Hare, J A Hartley, S McClean, L P Wakelin, L R Kelland, D J Robins
The aim of this work was to develop new bifunctional alkylating agents which damage DNA in a selective manner. In order to extend our previously published work on conformationally restricted nitrogen mustards containing one piperidine ring, new bispiperidine derivatives were designed with varying lengths of carbon chain between the two rings and structure-activity relationships in these systems were studied. Thus samples of new bispiperidine salts 22-26 with chloromethyl groups at the 2-positions and a bridge between the two nitrogen atoms of 2-6 carbon atoms were synthesized. The analogous new bis(p-nitrophenylcarbamates) 17-21 were also prepared. The free bases were designed to be bifunctional alkylating agents via aziridinium ion formation with different distances between the two alkylating sites. The bispiperidines 22-24 were shown to alkylate guanines at the 7-position in the major groove of DNA more selectively than melphalan. The bispiperidine 22 with the shortest two carbon bridge was the most reactive but it was less cytotoxic than melphalan in a human colon carcinoma cell line (IC50 value approximately 30 microM) and in a human chronic myeloid leukaemia cell line (IC50 value approximately 12 microM). The most cytotoxic compound in the latter cell line was the carbamate 17, with an IC50 value of approximately 0.3 microM, and carbamates 17, 19 and 20 were most potent in a panel of human ovarian carcinoma cell lines. These compounds also showed circumvention of acquired cisplatin resistance in three paired cell lines. The carbamates 17-21, however, were less efficient at alkylating and cross-linking naked DNA than the bispiperidines 22-26.
{"title":"Synthesis of new bifunctional compounds which selectively alkylate guanines in DNA.","authors":"N D Henderson, S M Lacy, C C O'Hare, J A Hartley, S McClean, L P Wakelin, L R Kelland, D J Robins","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>The aim of this work was to develop new bifunctional alkylating agents which damage DNA in a selective manner. In order to extend our previously published work on conformationally restricted nitrogen mustards containing one piperidine ring, new bispiperidine derivatives were designed with varying lengths of carbon chain between the two rings and structure-activity relationships in these systems were studied. Thus samples of new bispiperidine salts 22-26 with chloromethyl groups at the 2-positions and a bridge between the two nitrogen atoms of 2-6 carbon atoms were synthesized. The analogous new bis(p-nitrophenylcarbamates) 17-21 were also prepared. The free bases were designed to be bifunctional alkylating agents via aziridinium ion formation with different distances between the two alkylating sites. The bispiperidines 22-24 were shown to alkylate guanines at the 7-position in the major groove of DNA more selectively than melphalan. The bispiperidine 22 with the shortest two carbon bridge was the most reactive but it was less cytotoxic than melphalan in a human colon carcinoma cell line (IC50 value approximately 30 microM) and in a human chronic myeloid leukaemia cell line (IC50 value approximately 12 microM). The most cytotoxic compound in the latter cell line was the carbamate 17, with an IC50 value of approximately 0.3 microM, and carbamates 17, 19 and 20 were most potent in a panel of human ovarian carcinoma cell lines. These compounds also showed circumvention of acquired cisplatin resistance in three paired cell lines. The carbamates 17-21, however, were less efficient at alkylating and cross-linking naked DNA than the bispiperidines 22-26.</p>","PeriodicalId":7927,"journal":{"name":"Anti-cancer drug design","volume":"13 7","pages":"749-68"},"PeriodicalIF":0.0,"publicationDate":"1998-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"20719553","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
We have previously found that adamantylmaleimide derivatives inhibited the growth of several cancer cell lines in vitro. In this study we examined the effect of adamantylmaleimide derivatives on the in vivo and in vitro growth of human gastric cancer cells. Experimental results showed that N-1-adamantylmaleimide (AMI) and N-1-(3,5-dimethyladamantyl)maleimide (DMAMI) exert modest growth inhibitory activities in vitro against five different cancer cell lines. In contrast, N-1-(3,5-dimethyl-adamantyl)maleamic acid (DMAMA), N-1-adamantylmaleamic acid (AMA) and N-1-adamantylsuccinimide (ASI) were virtually inactive. These results suggest that the double bond of N-substituted maleimide plays a prominent role in their antitumor activities. Further analysis with flow cytometry showed an accumulation of apoptotic SC-M1 cells after treatment with 3-10 microM AMI or 5-20 microM DMAMI for up to 72 h. DNA fragmentation by gel electrophoresis confirmed that AMI- and DMAMI- induced cytotoxicity led to cell apoptosis. In addition, scanning electron microscopy (SEM) showed that treating cells with AMI (> or = 10 microM) for 24 h, significantly changed the morphology of SC-M1 cells, i.e. they had an irregular flat shape and the cell membrane was porous. The AMI-induced morphological changes of the cell membrane may lead to apoptosis of SC-M1 cells. AMI-induced growth inhibition was observed in vivo using SCID mice bearing SC-M1 tumors. The AMI-induced growth inhibition of SC-M1 tumor was dose-dependent.
{"title":"In vitro and in vivo growth inhibition of cancer cells by adamantylmaleimide derivatives.","authors":"J J Wang, Y T Chern, T Y Liu, C W Chi","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>We have previously found that adamantylmaleimide derivatives inhibited the growth of several cancer cell lines in vitro. In this study we examined the effect of adamantylmaleimide derivatives on the in vivo and in vitro growth of human gastric cancer cells. Experimental results showed that N-1-adamantylmaleimide (AMI) and N-1-(3,5-dimethyladamantyl)maleimide (DMAMI) exert modest growth inhibitory activities in vitro against five different cancer cell lines. In contrast, N-1-(3,5-dimethyl-adamantyl)maleamic acid (DMAMA), N-1-adamantylmaleamic acid (AMA) and N-1-adamantylsuccinimide (ASI) were virtually inactive. These results suggest that the double bond of N-substituted maleimide plays a prominent role in their antitumor activities. Further analysis with flow cytometry showed an accumulation of apoptotic SC-M1 cells after treatment with 3-10 microM AMI or 5-20 microM DMAMI for up to 72 h. DNA fragmentation by gel electrophoresis confirmed that AMI- and DMAMI- induced cytotoxicity led to cell apoptosis. In addition, scanning electron microscopy (SEM) showed that treating cells with AMI (> or = 10 microM) for 24 h, significantly changed the morphology of SC-M1 cells, i.e. they had an irregular flat shape and the cell membrane was porous. The AMI-induced morphological changes of the cell membrane may lead to apoptosis of SC-M1 cells. AMI-induced growth inhibition was observed in vivo using SCID mice bearing SC-M1 tumors. The AMI-induced growth inhibition of SC-M1 tumor was dose-dependent.</p>","PeriodicalId":7927,"journal":{"name":"Anti-cancer drug design","volume":"13 7","pages":"779-96"},"PeriodicalIF":0.0,"publicationDate":"1998-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"20719555","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
S P Mackay, L Comoe, B Desoize, O Duval, J C Jardillier, R D Waigh
Some members of a series of 12-alkyloxy benzo[c]phenanthridines are potent inhibitors of the growth of P388 tumour cells in vitro, with a strong dependence on the nature of the 12-substituent. Analogues with a quaternary nitrogen in the side chain bind strongly to DNA but are less active against the tumour cells. The multi-drug-resistant cell line Pr8/22 shows less sensitivity to the new compounds. K562 Human leukaemia cells undergo differentiation in the presence of the benzo[c]phenanthridine derivatives with a structure-activity relationship which does not correlate well with potency against the P388 cell line.
{"title":"The effect of 12-alkoxy modification on the in vitro antileukaemic activity of N-methyl 2,3,8,9-tetramethoxybenzo[c]phenanthridinium salts.","authors":"S P Mackay, L Comoe, B Desoize, O Duval, J C Jardillier, R D Waigh","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>Some members of a series of 12-alkyloxy benzo[c]phenanthridines are potent inhibitors of the growth of P388 tumour cells in vitro, with a strong dependence on the nature of the 12-substituent. Analogues with a quaternary nitrogen in the side chain bind strongly to DNA but are less active against the tumour cells. The multi-drug-resistant cell line Pr8/22 shows less sensitivity to the new compounds. K562 Human leukaemia cells undergo differentiation in the presence of the benzo[c]phenanthridine derivatives with a structure-activity relationship which does not correlate well with potency against the P388 cell line.</p>","PeriodicalId":7927,"journal":{"name":"Anti-cancer drug design","volume":"13 7","pages":"797-813"},"PeriodicalIF":0.0,"publicationDate":"1998-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"20719556","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
J D Jiang, J Roboz, I Weisz, L Deng, L Ma, J F Holland, J G Bekesi
The four title compounds (not hitherto reported) were synthesized from 3-aminobenzoic acid through its trifluoroacetic acid-acid chloride derivative, reaction with urea and aminolytic deprotection to yield 3-aminobenzoylurea, followed by unconventional haloacetylation. Three key factors were found essential for antitumor activity: (i) the cytotoxic nature of the halogen: I > Br > Cl > F (ID90 0.014->10 microM); (ii) the position of the halogen: only the 3-position (meta) expressed relevant activity; and (iii) the presence of the urea group (1-position). The selectivity of the bromo and iodo compounds were higher than those of vinblastine and paclitaxel in terms of cytotoxicity (ID50 ratios in nonmalignant myocardial fibroblasts and CEM leukemia cells) and therapeutic indices (P338 leukemia bearing mice). Relevant mechanisms of bioactivity were mitotic arrest and apoptosis. Complete inhibition of microtubule assembly occurred in cell-free systems (at 2.8 versus 2.1 microM for vinblastine); in contrast to paclitaxel, the target compounds did not interfere with microtubule disassembly. The strong cancericidal and antimicrotubular activities of the bromine and iodine compounds justify further exploration of their potential in antineoplastic chemotherapy.
这四种标题化合物(迄今未报道)是由3-氨基苯甲酸通过其三氟乙酸-氯酸衍生物,与尿素反应和氨解脱保护制得3-氨基苯甲酰脲,然后进行非常规卤代乙酰化合成的。发现三个关键因素对抗肿瘤活性至关重要:(i)卤素的细胞毒性:i > Br > Cl > F (ID90 0.014->10微米);(ii)卤素的位置:只有3位(元)表达了相关活性;(iii)脲基(1位)的存在。在细胞毒性(非恶性心肌成纤维细胞和CEM白血病细胞的ID50比)和治疗指标(P338白血病小鼠)方面,溴和碘化合物的选择性高于长春花碱和紫杉醇。相关的生物活性机制为有丝分裂阻滞和细胞凋亡。在无细胞系统中,微管组装发生完全抑制(长春花碱为2.8微米,长春花碱为2.1微米);与紫杉醇相反,目标化合物不会干扰微管的分解。溴和碘化合物具有很强的杀癌和抗小管活性,值得进一步探索其在抗肿瘤化疗中的潜力。
{"title":"Synthesis, cancericidal and antimicrotubule activities of 3-(haloacetamido)-benzoylureas.","authors":"J D Jiang, J Roboz, I Weisz, L Deng, L Ma, J F Holland, J G Bekesi","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>The four title compounds (not hitherto reported) were synthesized from 3-aminobenzoic acid through its trifluoroacetic acid-acid chloride derivative, reaction with urea and aminolytic deprotection to yield 3-aminobenzoylurea, followed by unconventional haloacetylation. Three key factors were found essential for antitumor activity: (i) the cytotoxic nature of the halogen: I > Br > Cl > F (ID90 0.014->10 microM); (ii) the position of the halogen: only the 3-position (meta) expressed relevant activity; and (iii) the presence of the urea group (1-position). The selectivity of the bromo and iodo compounds were higher than those of vinblastine and paclitaxel in terms of cytotoxicity (ID50 ratios in nonmalignant myocardial fibroblasts and CEM leukemia cells) and therapeutic indices (P338 leukemia bearing mice). Relevant mechanisms of bioactivity were mitotic arrest and apoptosis. Complete inhibition of microtubule assembly occurred in cell-free systems (at 2.8 versus 2.1 microM for vinblastine); in contrast to paclitaxel, the target compounds did not interfere with microtubule disassembly. The strong cancericidal and antimicrotubular activities of the bromine and iodine compounds justify further exploration of their potential in antineoplastic chemotherapy.</p>","PeriodicalId":7927,"journal":{"name":"Anti-cancer drug design","volume":"13 7","pages":"735-47"},"PeriodicalIF":0.0,"publicationDate":"1998-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"20718999","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
R T Dorr, W Bellamy, J D Liddil, A Baker, K W Bair
A mechanism of action study was performed with 14 novel DNA binding agents characterized structurally as 2-(arylmethylamino)-1,3-propanediols (AMAPs). Correlations between 8226 myeloma cell colony formation and DNA damage were performed using soft agar colony-forming assays and alkaline elution filter techniques respectively. The frequency of double-stranded breaks (DSBs), single-stranded breaks (SSBs) and DNA-protein cross-links were compared with cell growth inhibitory potency. Highly potent AMAPs in the colony formation assays included 91U86, an N-methyl-5-benzo(c)carbazole derivative, 773U82, a 3-substituted fluoranthene derivative, and crisnatol (770U82), the 6-substituted chrysene derivative. There was a high frequency of SSBs and DSBs with many analogues, but only SSBs occurred in a concentration-dependent fashion. Using regression analysis, the degree of single-strand damage correlated with cytotoxic potency for the AMAPs, with an R-value of 0.57 (P = 0.04). By gel electrophoresis assays, three clinically tested AMAPs, crisnatol BW 770U82, BW 502U83 and BW 773U82, were shown to inhibit the decatenation of pBR 322 DNA by purified topoisomerase-II (TOPO-II) enzymes. These results suggest that while some active AMAPs, such as crisnatol (BW 770U82), BW 502U83 and BW 773U82, inhibit TOPO-II enzymes, leading to protein-associated SSBs, other mechanisms, which do not involve DNA strand damage, must also contribute to the cytotoxic effects of this class of antitumor compounds. Intercalation has been well documented for these drugs and this may explain some of the growth inhibitory activity of the AMAPs.
{"title":"Correlation of cytotoxicity and protein-associated DNA strand breaks for 2-(arylmethylamino)-1,3-propanediols.","authors":"R T Dorr, W Bellamy, J D Liddil, A Baker, K W Bair","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>A mechanism of action study was performed with 14 novel DNA binding agents characterized structurally as 2-(arylmethylamino)-1,3-propanediols (AMAPs). Correlations between 8226 myeloma cell colony formation and DNA damage were performed using soft agar colony-forming assays and alkaline elution filter techniques respectively. The frequency of double-stranded breaks (DSBs), single-stranded breaks (SSBs) and DNA-protein cross-links were compared with cell growth inhibitory potency. Highly potent AMAPs in the colony formation assays included 91U86, an N-methyl-5-benzo(c)carbazole derivative, 773U82, a 3-substituted fluoranthene derivative, and crisnatol (770U82), the 6-substituted chrysene derivative. There was a high frequency of SSBs and DSBs with many analogues, but only SSBs occurred in a concentration-dependent fashion. Using regression analysis, the degree of single-strand damage correlated with cytotoxic potency for the AMAPs, with an R-value of 0.57 (P = 0.04). By gel electrophoresis assays, three clinically tested AMAPs, crisnatol BW 770U82, BW 502U83 and BW 773U82, were shown to inhibit the decatenation of pBR 322 DNA by purified topoisomerase-II (TOPO-II) enzymes. These results suggest that while some active AMAPs, such as crisnatol (BW 770U82), BW 502U83 and BW 773U82, inhibit TOPO-II enzymes, leading to protein-associated SSBs, other mechanisms, which do not involve DNA strand damage, must also contribute to the cytotoxic effects of this class of antitumor compounds. Intercalation has been well documented for these drugs and this may explain some of the growth inhibitory activity of the AMAPs.</p>","PeriodicalId":7927,"journal":{"name":"Anti-cancer drug design","volume":"13 7","pages":"825-35"},"PeriodicalIF":0.0,"publicationDate":"1998-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"20719558","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hypoxia occurs to a variable extent in a vast majority of rodent and human solid tumors. It results from an inadequate and disorganized tumor vasculature, and hence an impaired oxygen delivery. A probe for the non-invasive detection of tumor hypoxia could find important utility in the selection of patients for therapy with bioreductive agents, anti-angiogenic/anti-vascular therapies and hypoxia-targeted gene therapy. In addition, tumor hypoxia has been shown to predict for treatment outcome following radio- or chemotherapy in human cancers, the underlying mechanism for which may involve hypoxia driving genetic instability and resulting tumor progression. Beyond oncology, utility can also be envisaged in stroke, ischemic heart disease, peripheral vascular disease, arthritis and other disorders. Design, validation, preclinical development and current status of a fluorinated 2-nitroimidazole, N-(2-hydroxy-3,3,3-trifluoropropyl)-2-(2-nitro-l-imidazolyl) acetamide (SR 4554, CRC 94/17), which has been rationally designed for the measurement of tumor hypoxia by magnetic resonance spectroscopy (MRS) and imaging (MRI), are reviewed. Application in positron emission tomography (PET) detection is also proposed. Design goals were: (i) a nitro group with appropriate redox potential for selective reduction and binding in hypoxic tumor cells; (ii) hydrophilic/hydrogen bonding character in the side chain to limit nervous tissue penetration and prevent neurotoxicity; and (iii) three equivalent fluorine atoms to enhance MRS/MRI detection, located in a metabolically stable position. Reduction of SR 4554 by mouse liver microsomes was dependent on oxygen content, with a half-maximal inhibition at 0.48 +/- 0.06%. SR 4554 underwent nitroreduction by hypoxic but not oxic tumor cells in vitro and electron energy loss spectroscopic analysis showed selective retention in the hypoxic regions of multicellular tumor spheroids. Pharmacokinetic design goals were met. In particular, low brain tissue concentrations were seen in contrast to excellent tumor levels, as measured by high performance liquid chromatography. The extent of this restricted entry to brain tumor was surprising given the overall octanol/water partition coefficient and was attributed to the hydrophilic/hydrogen bonding character of the side chain. Quantitative MRS was used to assess the retention of 19F signal in murine tumors and human tumor xenografts. The 19F retention index (FRI; ratio of 19F signal levels at 6 h relative to that at 45 min) ranged from 0.5 to 1.0 and 0.2 to 0.9 for murine tumors and human xenografts respectively. The correlation between SR 4554 retention and pO2 was not a linear one, but when FRI was > 0.5, the % pO2 < or = 5 mmHg was always > 60%, indicating that high FRI was associated with low levels of oxygenation. Finally, whole body 19F-MRI in mice demonstrated that SR 4554 and related metabolites localized mainly in tumor, liver and bladder regions. A selective MRS signal was
{"title":"Preclinical development and current status of the fluorinated 2-nitroimidazole hypoxia probe N-(2-hydroxy-3,3,3-trifluoropropyl)-2-(2-nitro-1-imidazolyl) acetamide (SR 4554, CRC 94/17): a non-invasive diagnostic probe for the measurement of tumor hypoxia by magnetic resonance spectroscopy and imaging, and by positron emission tomography.","authors":"E O Aboagye, A B Kelson, M Tracy, P Workman","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>Hypoxia occurs to a variable extent in a vast majority of rodent and human solid tumors. It results from an inadequate and disorganized tumor vasculature, and hence an impaired oxygen delivery. A probe for the non-invasive detection of tumor hypoxia could find important utility in the selection of patients for therapy with bioreductive agents, anti-angiogenic/anti-vascular therapies and hypoxia-targeted gene therapy. In addition, tumor hypoxia has been shown to predict for treatment outcome following radio- or chemotherapy in human cancers, the underlying mechanism for which may involve hypoxia driving genetic instability and resulting tumor progression. Beyond oncology, utility can also be envisaged in stroke, ischemic heart disease, peripheral vascular disease, arthritis and other disorders. Design, validation, preclinical development and current status of a fluorinated 2-nitroimidazole, N-(2-hydroxy-3,3,3-trifluoropropyl)-2-(2-nitro-l-imidazolyl) acetamide (SR 4554, CRC 94/17), which has been rationally designed for the measurement of tumor hypoxia by magnetic resonance spectroscopy (MRS) and imaging (MRI), are reviewed. Application in positron emission tomography (PET) detection is also proposed. Design goals were: (i) a nitro group with appropriate redox potential for selective reduction and binding in hypoxic tumor cells; (ii) hydrophilic/hydrogen bonding character in the side chain to limit nervous tissue penetration and prevent neurotoxicity; and (iii) three equivalent fluorine atoms to enhance MRS/MRI detection, located in a metabolically stable position. Reduction of SR 4554 by mouse liver microsomes was dependent on oxygen content, with a half-maximal inhibition at 0.48 +/- 0.06%. SR 4554 underwent nitroreduction by hypoxic but not oxic tumor cells in vitro and electron energy loss spectroscopic analysis showed selective retention in the hypoxic regions of multicellular tumor spheroids. Pharmacokinetic design goals were met. In particular, low brain tissue concentrations were seen in contrast to excellent tumor levels, as measured by high performance liquid chromatography. The extent of this restricted entry to brain tumor was surprising given the overall octanol/water partition coefficient and was attributed to the hydrophilic/hydrogen bonding character of the side chain. Quantitative MRS was used to assess the retention of 19F signal in murine tumors and human tumor xenografts. The 19F retention index (FRI; ratio of 19F signal levels at 6 h relative to that at 45 min) ranged from 0.5 to 1.0 and 0.2 to 0.9 for murine tumors and human xenografts respectively. The correlation between SR 4554 retention and pO2 was not a linear one, but when FRI was > 0.5, the % pO2 < or = 5 mmHg was always > 60%, indicating that high FRI was associated with low levels of oxygenation. Finally, whole body 19F-MRI in mice demonstrated that SR 4554 and related metabolites localized mainly in tumor, liver and bladder regions. A selective MRS signal was ","PeriodicalId":7927,"journal":{"name":"Anti-cancer drug design","volume":"13 6","pages":"703-30"},"PeriodicalIF":0.0,"publicationDate":"1998-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"20668517","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Altered efficacy and selectivity of tyrosine kinase inhibitors of the activated states of protein tyrosine kinases.","authors":"A Levitzki, F D Böhmer","doi":"","DOIUrl":"","url":null,"abstract":"","PeriodicalId":7927,"journal":{"name":"Anti-cancer drug design","volume":"13 6","pages":"731-4"},"PeriodicalIF":0.0,"publicationDate":"1998-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"20668518","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bioreductive drugs are designed to be activated by enzymatic reduction in hypoxic regions of tumours, but activation of these drugs is not always fully suppressed by oxygen in normal tissues. A further limitation is that bioreductive drug activation depends on suitable reductases being expressed in the hypoxic zone. This essay proposes an alternative approach in which prodrugs are reduced, and thereby activated, in hypoxic regions by ionizing radiation rather than by enzymes. This strategy is theoretically attractive, but design requirements for such radiation-activated cytotoxins are challenging. In particular, the reducing capacity of radiation at clinically relevant doses is small, which necessitates the development of prodrugs capable of releasing very potent cytotoxins efficiently in hypoxic tissue. It is shown that nitroarylmethyl quaternary (NMQ) salts possess many of the features required of a radiation-activated prodrug. In some heterocyclic NMQ compounds the cytotoxicity of the latent cytotoxic amine effector is suppressed by > 100-fold in the prodrug form, and the effector is released rapidly by fragmentation following reduction by a single electron. Appreciable cytotoxic activation of NMQ prodrugs can be achieved by irradiation at clinically relevant doses in anoxic plasma. Some of the further drug design challenges required to develop a clinical agent based on this approach are outlined.
{"title":"Radiation-activated prodrugs as hypoxia-selective cytotoxins: model studies with nitroarylmethyl quaternary salts.","authors":"W R Wilson, M Tercel, R F Anderson, W A Denny","doi":"","DOIUrl":"","url":null,"abstract":"<p><p>Bioreductive drugs are designed to be activated by enzymatic reduction in hypoxic regions of tumours, but activation of these drugs is not always fully suppressed by oxygen in normal tissues. A further limitation is that bioreductive drug activation depends on suitable reductases being expressed in the hypoxic zone. This essay proposes an alternative approach in which prodrugs are reduced, and thereby activated, in hypoxic regions by ionizing radiation rather than by enzymes. This strategy is theoretically attractive, but design requirements for such radiation-activated cytotoxins are challenging. In particular, the reducing capacity of radiation at clinically relevant doses is small, which necessitates the development of prodrugs capable of releasing very potent cytotoxins efficiently in hypoxic tissue. It is shown that nitroarylmethyl quaternary (NMQ) salts possess many of the features required of a radiation-activated prodrug. In some heterocyclic NMQ compounds the cytotoxicity of the latent cytotoxic amine effector is suppressed by > 100-fold in the prodrug form, and the effector is released rapidly by fragmentation following reduction by a single electron. Appreciable cytotoxic activation of NMQ prodrugs can be achieved by irradiation at clinically relevant doses in anoxic plasma. Some of the further drug design challenges required to develop a clinical agent based on this approach are outlined.</p>","PeriodicalId":7927,"journal":{"name":"Anti-cancer drug design","volume":"13 6","pages":"663-85"},"PeriodicalIF":0.0,"publicationDate":"1998-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"20669170","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}