Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 103(2): 201-8, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20588272

ABSTRACT

BACKGROUND: Metastases cause most cancer-related deaths. We investigated the use of hypoxia-selective cytotoxins as adjuvants to radiotherapy in the control of metastatic tumour growth. METHODS: The NLCQ-1, RB6145 and tirapazamine were assessed against the spontaneously metastasising KHT model. Subcutaneous KHT tumours (250 mm(3)) were irradiated with 25 Gy (single fraction) to control primary growth. Equitoxic drug treatments (NLCQ-1 (10 mg kg(-1)) once daily; RB6145 (75 mg kg(-1)) and tirapazamine (13 mg kg(-1)) twice daily) were administered 3-6 days post-radiotherapy when hypoxic cells were evident in lung micrometastases. Mice were culled when 50% of controls exhibited detrimental signs of lung metastases. RESULTS: In total, 95% of control mice presented with lung disease. This was significantly reduced by NLCQ-1 (33%; P=0.0002) and RB6145 (60%; P=0.02). Semi-quantitative grading of lung disease revealed a significant improvement with all treatments, with NLCQ-1 proving most efficacious (median grades: control, 4; NLCQ, 0 (P<0.0001); RB6145, 1 (P<0.001), tirapazamine, 3 (P=0.007)). Positron emission tomography (PET) was evaluated as a non-invasive means of assessing metastatic development. Primary and metastatic KHT tumours showed robust uptake of [(18)F]fluorodeoxyglucose ([(18)F]FDG). Metastatic burden discernable by [(18)F]FDG PET correlated well with macroscopic and histological lung analysis. CONCLUSION: The hypoxia-selective cytotoxin NLCQ-1 controls metastatic disease and may be a successful adjuvant to radiotherapy in the clinical setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Hypoxia/drug effects , Imidazoles/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Quinolines/administration & dosage , Sarcoma/drug therapy , Sarcoma/secondary , Animals , Cell Line, Tumor , Chemotherapy, Adjuvant , Combined Modality Therapy , Drug Administration Schedule , Drug Evaluation, Preclinical , Mice , Mice, Inbred C3H , Neoplasm Metastasis , Nitroimidazoles/administration & dosage , Tirapazamine , Triazines/administration & dosage
2.
Br J Radiol ; 81 Spec No 1: S45-56, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18819998

ABSTRACT

Hypoxia, or a lack of oxygen, occurs in 50-60% of solid human tumours. Clinical studies have shown that the presence and extent of hypoxia in a tumour cannot be predicted by size or histopathological stage but it is predictive of a poor outcome following radiotherapy, chemotherapy and surgery. However, as a physiological feature of tumours, it can be exploited and researchers have developed many hypoxia-selective chemotherapies or bioreductive drugs that are in varying stages of clinical development. These agents are prodrugs that have two key requirements for their biological activation: they require the reductive environment of a hypoxic tumour cell and the appropriate complement of cellular reductase enzymes. To overcome tumour heterogeneity in reductase enzyme levels and enhance bioreductive drug metabolism a gene therapy strategy can be employed. We have reviewed this field and also present our own pre-clinical research using gene therapy to enhance bioreductive drug treatment for the treatment of cancer. We have specifically focused on studies enhancing lead clinical bioreductive drugs. We consider the metabolic requirements for their activation and we highlight the key in vivo studies supporting the future clinical development of hypoxia-targeted gene-directed enzyme prodrug therapy.


Subject(s)
Hypoxia/therapy , Neoplasms/therapy , Prodrugs/therapeutic use , Alkylating Agents/metabolism , Alkylating Agents/therapeutic use , Animals , Anthraquinones , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/therapeutic use , Cytochromes/metabolism , Cytochromes/therapeutic use , Cytochromes b5/metabolism , Cytochromes b5/therapeutic use , Female , Genetic Therapy , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1/metabolism , Mice , Mitomycin/metabolism , Mitomycin/therapeutic use , Neoplasms/genetics , Neoplasms/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase/therapeutic use , Prodrugs/metabolism , Xanthine Oxidase/metabolism , Xanthine Oxidase/therapeutic use
3.
Oncol Res ; 15(4): 219-31, 2005.
Article in English | MEDLINE | ID: mdl-17822282

ABSTRACT

Nitro(imidazole/triazole)-linked acridines (NLAs) have been previously developed in our laboratory as DNA-intercalating bioreductive drugs. Such compounds demonstrate toxicity through the formation of bulky monoadducts with cellular macromolecules upon activation and reductive metabolism under hypoxic conditions. However, NLAs also demonstrate considerable aerobic toxicity. Based on the ability of NLAs to bind strongly to DNA through intercalation, we investigated whether their relatively high aerobic cytotoxicity and their relatively low hypoxic selectivity in vitro are associated with topoisomerases I and II (Topo I and II) inhibition. DNA Topo I or II-mediated activity studies have been performed using supercoiled or kinetoplast DNA plasmids. Calf thymus or human Topo I and human Topo II purified enzymes were used. All NLA derivatives strongly inhibited relaxation of supercoiled DNA catalyzed by either Topo I or II, in a concentration-dependent manner, without stabilization of a cleavable complex. Aerobic toxicity correlated well with the inhibition of Topo II-mediated decatenation of kinetoplast DNA, whereas the intracellular concentrations of NLAs were 27-152-fold greater than those needed for 50% inhibition of Topo-II mediated decatenation of DNA. These results suggest that topoisomerase inhibition accounts for NLAs aerobic toxicity.


Subject(s)
Acridines/pharmacology , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type I/metabolism , DNA, Kinetoplast/drug effects , DNA, Superhelical/drug effects , Intercalating Agents/pharmacology , Acridines/chemistry , Aerobiosis , Animals , Cattle , Cell Hypoxia , Cell Line , Cricetinae , Cricetulus , DNA, Kinetoplast/metabolism , DNA, Superhelical/metabolism , DNA-Binding Proteins/metabolism , Humans , Intercalating Agents/chemistry , Molecular Structure , Plasmids , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors
4.
Oncol Res ; 13(12): 561-6, 2003.
Article in English | MEDLINE | ID: mdl-12899246

ABSTRACT

The antitumor effect of cyclophosphamide (CPM) and paclitaxel was investigated in BALB/c mice bearing EMT6 tumors, in combination with the bioreductive compound NLCPQ-1 by using the in vivo/in vitro assay as the endpoint. An optimum administration schedule for a synergistic interaction between NLCPQ-1 and CPM/paclitaxel was determined and dose modification factors (DMF) were calculated for antitumor effect and bone marrow toxicity. All drugs were given by IP injection; NLCPQ-1 at 15 mg/kg, which is much less than its maximally tolerated dose (MTD greater than 50 mg/kg), paclitaxel up to 25 mg/kg, and CPM up to 200 mg/kg. Bone marrow toxicity studies were performed in parallel by using a modified CFU-GM assay. A schedule-dependent synergistic interaction was observed for both chemotherapeutic agents combined with NLCPQ-1 but with entirely different patterns, as has been previously seen with the analog NLCQ-1. The optimal degree of potentiation, P (percentage of tumor cells that were killed due to clear potentiation), was 31 and 33 when NLCPQ-1 was administered 2 h before CPM and 3-3.5 h after paclitaxel, respectively. At the above time schedules, NLCPQ-1 modified the dose of CPM and paclitaxel, for 60% tumor cell killing, by a factor of 1.8 and 2.1, respectively. Bone marrow toxicity was not enhanced by combining either chemotherapeutic agent with NLCPQ-1. Comparison with results from previous similar studies with NLCQ-1 revealed that, on a molar basis, NLCPQ-1 was a less potent chemosensitizer than NLCQ-1. However, the results still suggest a potential clinical use of NLCPQ-1 as an adjuvant to CPM or paclitaxel therapy against solid tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cyclophosphamide/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Nitroimidazoles/pharmacology , Paclitaxel/pharmacology , Quinolines/pharmacology , Animals , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cyclophosphamide/administration & dosage , Drug Administration Schedule , Drug Synergism , Female , Mice , Mice, Inbred BALB C , Nitroimidazoles/administration & dosage , Paclitaxel/administration & dosage , Quinolines/administration & dosage , Tumor Cells, Cultured
5.
Cancer Chemother Pharmacol ; 50(4): 291-8, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12357303

ABSTRACT

PURPOSE: The antitumor effect and bone marrow toxicity of 5-fluorouracil (5FU) in combination with the hypoxia-selective cytotoxins NLCQ-1 or tirapazamine (TPZ) were investigated in vivo. METHODS: Using appropriate intraperitoneal administration schedules for optimal synergistic interactions, the antitumor effect and the bone marrow toxicity of combinations of NLCQ-1 or TPZ and 5FU were determined in EMT6/BALB/c and SCCVII/C3H models in terms of dose modification factors (DMF) using the in vivo-in vitro clonogenic assay as endpoint. Bone marrow toxicity studies were performed in parallel using a modified CFU-GM assay. The antitumor efficacies of each combination treatment under optimal administration conditions were evaluated in the SCCVII/C3H model using also the tumor regrowth assay as endpoint. RESULTS: A schedule-dependent and tumor-specific synergistic interaction was observed for NLCQ-1 plus 5FU and DMFs of 2.0-2.3 and 1.0 were obtained for the antitumor effect and bone marrow toxicity, respectively, in both tumor models. The antitumor effect of 5FU was slightly potentiated (DMF 1.2) by TPZ in the EMT6/BALB/c model but not in the SCCVII/C3H model when the in vivo-in vitro assay was used as the endpoint. Significant additional tumor regrowth delays (about 11 and 6 days for NLCQ-1 and TPZ, respectively) were observed, compared to the effect of 5FU alone, when an equitoxic dose of NLCQ-1 (10 mg/kg) or TPZ (23 mg/kg) was administered 1 h before 5FU (50 mg/kg) twice a day at 4-h intervals on days 0 and 9. CONCLUSIONS: These results corroborate the therapeutic advantage of combining hypoxia-selective cytotoxins such as NLCQ-1 and TPZ with chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Fluorouracil/administration & dosage , Hypoxia/drug therapy , Imidazoles/administration & dosage , Quinolines/administration & dosage , Triazines/administration & dosage , Animals , Bone Marrow/drug effects , Drug Synergism , Female , Fluorouracil/adverse effects , Imidazoles/adverse effects , Mice , Mice, Inbred BALB C , Quinolines/adverse effects , Tirapazamine , Triazines/adverse effects , Tumor Cells, Cultured , Tumor Stem Cell Assay
6.
In Vivo ; 15(5): 365-71, 2001.
Article in English | MEDLINE | ID: mdl-11695231

ABSTRACT

19F-labeled bioreductive drugs bound to hypoxic cells in tumors could be detected by nuclear magnetic resonance, provided that they do not lose 19F during their metabolism. NLTQ-1, a 2-nitroimidazole-linked 7-trifluoromethylquinoline, has been synthesized to furnish this aim. NLTQ-1 demonstrated hypoxic selectivities of 7-10 in various cell-lines, in vitro. Uptake studies in V79 cells showed a 5 to 6 fold greater intracellular than extracellular concentration at a range of 100-300 microM input concentrations. A strong sharp peak, which was identified as the parent compound, was observed in the 19F-NMR spectrum of 90% MeCN extracts of V79 cells aerobically exposed to NLTQ-1, indicating that NLTQ-1 was not metabolized under aerobic conditions. Similarly, 19F NMR efflux studies in intact cells showed that the NLTQ-1 was bound to the cells predominantly under hypoxic conditions. 19F-NMR spectra of intact cells, exposed under hypoxic conditions to NLTQ-1, and of their lysates, after precipitation of various cellular components, indicated that possible covalent binding of NLTQ-1 had occurred with macromolecules such as proteins and nucleic acids. Therefore, NLTQ-1 might be suitable as a 19F-MRS/MRI hypoxia probe, although further in vivo work is necessary to verify this matter.


Subject(s)
Cell Hypoxia , Fluorine/analysis , Imidazoles/analysis , Magnetic Resonance Spectroscopy , Quinolines/analysis , Reducing Agents/analysis , Aerobiosis , Animals , Cell Fractionation , Cell Line/chemistry , Cell Line/drug effects , Cricetinae , Cricetulus , DNA/metabolism , Extracellular Space/chemistry , Female , Fibroblasts/chemistry , Fibroblasts/drug effects , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Imidazoles/toxicity , Intracellular Fluid/chemistry , Isotopes/analysis , Lung/cytology , Molecular Structure , Ovarian Neoplasms/pathology , Protein Binding , Quinolines/chemistry , Quinolines/metabolism , Quinolines/toxicity , Reducing Agents/chemistry , Reducing Agents/metabolism , Reducing Agents/toxicity , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/drug effects
7.
Oncol Res ; 12(8): 325-33, 2001.
Article in English | MEDLINE | ID: mdl-11589303

ABSTRACT

The novel hypoxia-selective cytotoxin NLCQ-1, which is a weak DNA intercalator, was studied in conjunction with radiation against V79 cultured cells and EMT6 or SCCVII tumors in their syngeneic mice and compared with tirapazamine (TPZ). NLCQ-1 was a very potent and efficient radiosensitizer of hypoxic V79 cells, providing SER values of 2.27-2.56 at 20-80 microM concentration (measured at 10% survival level). Its C1.6 (concentration for an SER of 1.6 to be obtained) was 7.2+/-0.2 microM. Its in vitro therapeutic index (ThI, defined as CT50(Air),/C1.6) varied by the exposure time from 57 (1-h exposure) to 145 (4.5-h exposure). The corresponding C1.6 value for TPZ was 16.9 microM whereas its in vitro therapeutic index was 49 (3-h exposure). A schedule-dependent synergistic interaction was observed between NLCQ-1 or TPZ and 20 Gy of radiation in both tumor models examined, by using the in vivo-in vitro assay as endpoint. Optimal synergism (> 1 log) was observed in EMT6 tumors when each bioreductive drug was given between 45 and 60 min before irradiation. NLCQ-1 alone had no significant antitumor activity at 10 mg/kg (28% of its single LD50), whereas a 0.4 surviving fraction was obtained by TPZ at 30 mg/kg (38% of its single LD50). SER values of 1.52 and 1.25 were obtained with 10 mg/kg NLCQ-1 and 30 mg/kg TPZ, respectively, in EMT6 tumors. An SER value of 1.58 was obtained for both hypoxia-selective cytotoxins, at equitoxic doses, in SCCVII tumors, by using a fractionated regimen. These results suggest a possible use of NLCQ-1 or TPZ as adjuvants to radiotherapy.


Subject(s)
Antineoplastic Agents/toxicity , Imidazoles/toxicity , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/radiotherapy , Quinolines/toxicity , Radiation-Sensitizing Agents/toxicity , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/radiotherapy , Cell Hypoxia , Cell Line , Cell Survival , Cricetinae , Cytotoxins/administration & dosage , Cytotoxins/adverse effects , Cytotoxins/metabolism , Cytotoxins/pharmacology , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Administration Schedule , Female , Imidazoles/administration & dosage , Imidazoles/metabolism , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/radiotherapy , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Neoplasms, Experimental/pathology , Quinolines/administration & dosage , Quinolines/metabolism , Radiation-Sensitizing Agents/administration & dosage , Radiation-Sensitizing Agents/metabolism , Tirapazamine , Triazines/administration & dosage , Triazines/metabolism , Triazines/toxicity , Tumor Cells, Cultured , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
8.
Cancer Chemother Pharmacol ; 48(2): 160-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11561782

ABSTRACT

PURPOSE: Comparisons of schedule-dependent interactions between the hypoxic cytotoxins NLCQ-1/ tirapazamine (TPZ) and various chemotherapeutic drugs in BALB/c mice bearing EMT6 tumors. METHODS: The antitumor effects of the single or combined drugs were assessed with various administration time intervals using the in vivo-in vitro clonogenic assay as the endpoint. The chemotherapeutic drugs tested were cisplatin (cisDDP), melphalan (L-PAM), cyclophosphamide (CPM), 5-fluorouracil (5-FU), doxorubicin (Doxo), etoposide (VP-16) and Taxol at doses of 8, 5, 100, 150, 12, 35 and 20 mg/kg, respectively. NLCQ-1 was given at 10 mg/kg (28% of its single LD50 value) and TPZ was given at 30 mg/kg (38% of its single LD50 value). All drugs were given by i.p. injection in saline or as commercially available pharmaceutical solutions. RESULTS: Schedule-dependent synergistic interactions with different patterns for each bioreductive drug were observed with almost all of the chemotherapeutic agents examined. Potentiation accounting for more than 25% of the total tumor cell killing was observed with NLCQ-1/TPZ and cisDDP, L-PAM, CPM, 5-FU and Taxol at the optimal administration intervals. Potentiation accounting for 70% of the total tumor cell killing was found with NLCQ-1 and CPM. CONCLUSIONS: These results suggest a potential clinical use of NLCQ-1/TPZ as adjuvants to certain chemotherapeutic agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Imidazoles/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Quinolines/pharmacology , Triazines/pharmacology , Animals , Cisplatin/administration & dosage , Cisplatin/pharmacology , Cyclophosphamide/administration & dosage , Cyclophosphamide/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Administration Schedule , Drug Synergism , Etoposide/administration & dosage , Etoposide/pharmacology , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Imidazoles/administration & dosage , Melphalan/administration & dosage , Melphalan/pharmacology , Mice , Mice, Inbred BALB C , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Quinolines/administration & dosage , Tirapazamine , Triazines/administration & dosage , Tumor Cells, Cultured
9.
Oncol Res ; 12(4): 185-92, 2000.
Article in English | MEDLINE | ID: mdl-11341468

ABSTRACT

A novel weakly DNA-intercalative bioreductive compound. 4-[3-(2-nitro-1-imidazolyl)-propylamino]-7-chloroquinoline hydrochloride (NLCQ-1). has been synthesized and studied as a hypoxia-selective cytotoxin in vitro. NLCQ-1, which shares a similar structure with the DNA-intercalative antimalarial drug chloroquine, bound more strongly to DNA than the nonchlorinated analog NLQ-1 (4-[3-(2-nitro-1-imidazolyl)propylamino]-quinaldine hydrochloride). Thus, NLCQ-1 exhibited a C50 [concentration for 50% displacement of the ethidium bromide (EB) from a DNA-EB complex] of 44 microM, whereas a C50 value could not be reached for NLQ-1 up to 225 microM. NLCQ-1 demonstrated significant hypoxic selectivity in several rodent (V79, EMT6, SCCVII) or human (A549, OVCAR-3) tumor cell lines. Its potency as a hypoxic cytotoxin (expressed as the product of exposure time and concentration for 50% survival) ranged between 10 and 136 microM x h, for the cell lines tested, at 30 microM input concentration. Because uptake in all cell lines was similar, the differences in potency may reflect differences in the enzymatic profile or damage repair processes among the cell lines. In addition, however, the most striking feature of NLCQ-1 was that hypoxic selectivity increased with exposure time, a common feature normally found in only bis-bioreductive agents carrying two moieties with different redox potentials. Thus, hypoxic selectivity of NLCQ-1 in V79 cells at 50% survival was increased from fivefold up to 388-fold by increasing exposure time from 1 to 4.5 h, as the result of a concomitant increase and decrease in its hypoxic and aerobic potency, respectively, over time. Because the nonchlorinated analog NLQ-1 did not demonstrate similar behavior, we hypothesized that the C-7 chlorine of NLCQ-1 might play a significant role in this phenomenon.


Subject(s)
Aminoquinolines/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Hypoxia , Imidazoles/therapeutic use , Quinolines/therapeutic use , Animals , Cell Survival , Chloroquine/chemistry , Chromatography, Thin Layer , Cytotoxins , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Models, Chemical , Neoplasms/drug therapy , Rats , Time Factors , Tumor Cells, Cultured
10.
Oncol Res ; 11(8): 345-57, 1999.
Article in English | MEDLINE | ID: mdl-10803738

ABSTRACT

9-[3-(2-Nitro-1-imidazolyl)propylamino]-1,2,3,4-tetrahydroacridine hydrochloride (THNLA-1) is a 2-nitroimidazole-based, weakly DNA-intercalating bioreductive agent that significantly potentiates the toxic effects of commonly used antitumor drugs such as melphalan (L-PAM) or cis-DDP in sensitive or resistant cell lines in culture, as well as in solid tumors in mice. Potentiation in vitro was observed when cells were preexposed to THNLA-1 under hypoxic conditions before exposure to L-PAM under aerobic conditions. In this study we investigated possible mechanisms involved in the potentiation of L-PAM by THNLA-1 in V79 Chinese hamster cells. Limited depletion of glutathione with buthionine sulfoximine or THNLA-1 under hypoxic pretreatment conditions accounted for only 8.3% of the potentiation induced by THNLA-1. However, DNA, RNA, and protein synthesis were inhibited in a synergistic way in cells preexposed to THNLA-1 under hypoxic conditions (2 h, 37 degrees C) and then coexposed to various doses of L-PAM under aerobic conditions (1 h, 37 degrees C). Cell cycle analysis by flow cytometry showed a slow traverse through the S phase in the L-PAM-alone-treated cells. However, this phenomenon was more prominent in the THNLA-1 plus L-PAM-treated cells. Under aerobic co-incubation conditions with L-PAM, no difference was observed in the cell cycle of L-PAM-alone-treated cells vs. THNLA-1 plus L-PAM-treated cells. Significantly increased apoptosis was observed in the hypoxia-pretreated cells with THNLA-1, 12 and 24 h posttreatment. Comet and alkaline elution assay analysis showed increased DNA cross-links in the hypoxia-pretreated cells with THNLA-1 compared to the L-PAM-alone-treated cells. Finally, potential lethal damage repair was totally suppressed only in the hypoxia-pretreated cells with THNLA-1. In conclusion, DNA damage and hindrance in its repair are the most important mechanisms in the potentiation of L-PAM by THNLA-1, under hypoxic pretreatment conditions.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Melphalan/pharmacology , Radiation-Sensitizing Agents/pharmacology , Tacrine/analogs & derivatives , Animals , Apoptosis/drug effects , Buthionine Sulfoximine/pharmacology , Cell Cycle/drug effects , Cell Hypoxia/drug effects , Cell Line/drug effects , Comet Assay , Cricetinae , Cricetulus , DNA/biosynthesis , DNA/drug effects , DNA Damage/drug effects , DNA Repair/drug effects , Drug Synergism , Glutathione/metabolism , Protein Biosynthesis , Proteins/drug effects , RNA/biosynthesis , RNA/drug effects , Tacrine/pharmacology
11.
Int J Radiat Oncol Biol Phys ; 42(4): 775-9, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9845094

ABSTRACT

PURPOSE: To investigate in vivo interactions between the recently developed bioreductive agent 4-[3-(2-nitroimidazolyl)-propylamino]-7-chloroquinoline hydrochloride (NLCQ-1) and the chemotherapeutic agents melphalan (L-PAM), cis-platin (cisDDP) and cyclophosphamide (CPM). METHODS AND MATERIALS: EMT6 and FSaIIC tumor cells were inoculated (subcutaneously) into the leg(s) of female Balb/c and male C3H mice, respectively. Treatment was initiated at 10 mm (EMT6) and 5 mm (FSaIIC) mean tumor diameter. The in vivo-in vitro and tumor regrowth assays were used, respectively, as endpoints. Bone marrow toxicity studies were also performed when the in vivo-in vitro assay was used. Drugs were given by i.p. injection. Tumors were excised 18-h after chemotherapeutic drug administration (Balb/c mice) or measured daily until three times their original size (C3H mice). The optimum administration schedule for potentiation between NLCQ-1 and each chemotherapeutic drug, as well as dose modification factors (DMF) at the optimum time, were determined with the in vivo-in vitro assay. When the tumor regrowth assay was used, each chemotherapeutic agent was given either as a single dose or as a split dose over two consecutive days at the optimum administration time after a 10 mg/kg NLCQ-1 i.p. injection. RESULTS: NLCQ-1 (at 0.33 times MTD), strongly potentiated the antitumor effect of L-PAM, cisDDP and CPM without concurrent enhancement in bone marrow toxicity. Potentiation was strictly schedule dependent and the optimum effect (1.5 to 2 logs killing beyond additivity) was observed when NLCQ-1 was given 60-, 45-, and 110-min before L-PAM, cisDDP, and CPM, respectively. The DMF values at 30% survival were 2.5, 1.9, and 3.8 for L-PAM, cisDDP, and CPM, respectively. DMF values for bone marrow toxicity at 50% survival were ca. 1 for all chemotherapeutic drugs. Pretreatment with NLCQ-1 resulted in 4-12 days extra delay in the regrowth of FSaIIC tumors. CONCLUSIONS: These results support the clinical investigation of NLCQ-1 as a chemosensitizer.


Subject(s)
Aminoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Prodrugs/pharmacology , Animals , Cisplatin/pharmacology , Cyclophosphamide/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Female , Male , Melphalan/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H
12.
Oncol Res ; 10(3): 163-73, 1998.
Article in English | MEDLINE | ID: mdl-9700727

ABSTRACT

NLCPQ-1 is a novel, weak, DNA-affinic bioreductive compound with enhanced chemosensitizing ability for commonly used chemotherapeutic agents, both in vitro and in vivo. In the present report we investigated possible mechanisms involved in the potentiation of cis-DDP and L-PAM in V79 cells. Potentiation was observed when cells were pretreated under hypoxic conditions with NLCPQ-1 prior to their aerobic exposure to each chemotherapeutic agent studied and in the presence of NLCPQ-1. The dominant mechanisms, under hypoxic pretreatment conditions, participating in the potentiation were: a) extensive DNA damage, as measured by the comet and alkaline elution assays, b) DNA, RNA, and protein synthesis inhibition, c) significant delay in the traverse through the S phase, as observed by flow cytometry, and possibly d) suppression of PLD repair. Apoptosis was also detected 36 h posttreatment in the chemotherapeutic drug-treated as well as the combination drug-treated cells. Glutathione depletion by NLCPQ-1 metabolites under hypoxic conditions was also involved in the potentiation process, but its contribution in potentiation was minimal.


Subject(s)
Antineoplastic Agents/metabolism , DNA/metabolism , Nitroimidazoles/metabolism , Quinolines/metabolism , Alkylating Agents/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Hypoxia/drug effects , Cell Line , Cricetinae , Cricetulus , DNA/antagonists & inhibitors , DNA/biosynthesis , Flow Cytometry , Glutathione/metabolism , Microscopy, Fluorescence , Nitroimidazoles/pharmacology , Protein Synthesis Inhibitors/metabolism , Protein Synthesis Inhibitors/pharmacology , Quinolines/pharmacology , RNA/antagonists & inhibitors , RNA/biosynthesis
13.
Oncol Res ; 9(5): 249-57, 1997.
Article in English | MEDLINE | ID: mdl-9306432

ABSTRACT

9-(3-(2-Nitro-1-imidazolyl)propylamino]-cyclopentano[b]quinoline hydrochloride (NLCPQ-1) is one member of a limited series of 2-nitroimidazole-linked derivatized quinolines we have synthesized to be weak DNA binding compounds. On a concentration basis, NLCPQ-1 is the most potent analogue of the series as a radiosensitizer and cytotoxin of hypoxic cells in vitro and in vivo. This improved efficacy compared to untargeted nitroimidazolic bioreductive compounds has been mainly attributed to its weak DNA binding. In the present study, we investigated the ability of NLCPQ-1 to act synergistically with 4-[bis(2-chloroethyl)amino]-L-phenylalanine (L-PAM, melphalan) or cis-diamminedichloroplatinum (cis-DDP, cisplatin) against tumor cells in vitro and in vivo. We demonstrated that 7 microM NLCPQ-1 potentiated the toxic effect of cis-DDP and L-PAM against V79 cells under hypoxic pretreatment conditions with dose modification factors (DMF) of 2.6 and 2.4, respectively, measured at 0.1 survival. Potentiation was dependent on the concentrations of both the chemotherapeutic agent and NLCPQ-1 as well as on the duration of the hypoxic pretreatment with NLCPQ-1. No potentiation was observed under aerobic cotreatment conditions in vitro. Significant synergism was observed when 15 mg/kg NLCPQ-1 was administered IP at various time intervals before a single dose of L-PAM (5 mg/kg) or cis-DDP (5 mg/kg) in Balb/c mice bearing EMT6 tumors. The in vivo/in vitro assay was used as the investigational endpoint and either the fractional product or isobologramic analysis was used to determine synergistic interactions.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/toxicity , DNA, Neoplasm/drug effects , Melphalan/toxicity , Nitroimidazoles/pharmacology , Quinolines/pharmacology , Animals , Antineoplastic Agents/metabolism , Cell Survival/drug effects , Cells, Cultured , Cricetinae , Cricetulus , DNA Adducts/drug effects , Drug Synergism , Female , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Nitroimidazoles/metabolism , Quinolines/metabolism , Tumor Cells, Cultured/drug effects
14.
Br J Cancer Suppl ; 27: S267-70, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8763895

ABSTRACT

THNLA-1 contains a 2-nitroimidazole tethered to 9-amino-1,2,3,4-tetrahydroacridine. Compared with its parent acridinic analogue, NLA-1, THNLA-1 is a weak DNA-affinic bioreductive compound with a greater mobility along the DNA backbone, decreased aerobic toxicity, greater hypoxic selectivity and a superior in vitro therapeutic index. Also, THNLA-1 behaves as a radio/chemosensitiser in vitro. In this report we have expanded our radio/chemosensitisation studies in vivo, using the EMT-6 mouse mammary tumour model in balb/c mice and the in vivo-in vitro assay. THNLA-1 was given i.p. ( < or = 0.5 ml in saline) at various time intervals before a single dose of 20 Gy whole-body irradiation. Tumours were excised immediately or 24 h after irradiation. Radiosensitisation studies with SR-2508 (i.v.) have been performed in a similar way for comparison purposes. THNLA-1 demonstrated the same radiosensitising effect as SR-2508 but with 19-fold less dose (mmol kg-1). The optimum effect was observed when THNLA-1 was given 1 h before irradiation and the tumours excised 24 h after irradiation. Chemosensitisation studies in the same tumour model and using cis DDP showed that the cytotoxic effect of cis-DDP (5 or 8 mg kg-1, i.p.) was significantly enhanced with 30 or 45 mg kg-1 THNLA-1 given approximately 3 h before cis-DDP. A similar potentiating effect was observed when NLA-1 (27 or 30 mg kg-1) was used, but toxicity was also observed at the higher dose. Limited toxicity studies showed that THNLA-1 is well tolerated up to at least 70 mg kg-1 as a single dose, for more than 40 days.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mammary Neoplasms, Experimental/therapy , Radiation-Sensitizing Agents/pharmacology , Tacrine/analogs & derivatives , Animals , Cisplatin/administration & dosage , Drug Synergism , Etanidazole/pharmacology , Female , Mice , Mice, Inbred BALB C , Tacrine/administration & dosage , Tacrine/pharmacology
15.
In Vivo ; 10(1): 49-57, 1996.
Article in English | MEDLINE | ID: mdl-8726811

ABSTRACT

THNLA-1 is a recently synthesized 2-nitroimidazole based, DNA-affinic bioreductive agent. It features a tetrahydroacridinic chromophore, which allows loose binding to DNA and therefore greater mobility along its backbone. THNLA-1 was proved to be a very good radiosensitizer and cytotoxin of hypoxic cells in vitro with an improved therapeutic index compared to the fully aromatic analog NLA-1. In this report we investigated the interaction of THNLA-1 with cis-DDP or L-PAM in the sensitive V79 and resistant OVCAR cells, using various schedule protocols. Also, the THNLA-1/cis-DDP interaction in balb/c mice has been investigated, using the EMT-6 mouse tumors. Isobologramic as well as fractional product concept analysis, clearly showed that synergistic interaction occurs between THNLA-1 and each chemotherapeutic agent, under hypoxic pretreatment conditions of the cells with THNLA-1 in vitro. The dose modification factor (DMF) values obtained in the resistant OVCAR-3 cells are similar to those obtained for the approximately 4 times more sensitive V79 cells. Therefore, the DMF value for e.g. L-PAM at 0.1 survival fraction, is approximately 2.76 when 15 microM THNLA-1 was used in OVCAR-3 cells, and approximately 2.50, when 10 microM THNLA-1 was used in V79. The supra-additive effect is dependent on the hypoxia-pretreatment time with THNLA-1, on THNLA-1 concentration and on the concentration of the chemotherapeutic drug. The limited in vivo study showed that THNLA-1, at doses significantly lower than its MTD, strongly potentiates the killing effect of cis-DDP and that the optimum effect during the combination treatment was observed when THNLA-1 was administered i.p., 2.5-3.0 h before cis-DDP. Toxicity studies in balb/c mice (without tumors) showed that THNLA-1 is well tolerated up to at least 70 mg/kg for more than 40 days while no toxicity was observed with the combined drugs used in our experimental protocol. These results are promising for the potential clinical use of THNLA-1 as an adjuvant in chemotherapy.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cisplatin/pharmacology , DNA/drug effects , Melphalan/pharmacology , Radiation-Sensitizing Agents/pharmacology , Tacrine/analogs & derivatives , Animals , Cell Hypoxia , Cell Line , Cell Survival , Cricetinae , Drug Interactions , Female , Mice , Mice, Inbred BALB C , Molecular Structure , Radiation-Sensitizing Agents/toxicity , Tacrine/pharmacology , Tacrine/toxicity , Time Factors , Tumor Cells, Cultured
16.
Oncol Res ; 8(10-11): 425-34, 1996.
Article in English | MEDLINE | ID: mdl-9114435

ABSTRACT

Our recent studies with 9-[3-(2-nitro-1-imidazolyl)propylamino]-1,2,3,4,-tetrahydroacridine++ + hydrochloride (a 2-nitroimidazole-linked 1,2,3,4-tetrahydroacridine derivative) shows that less effective DNA binding indeed leads to a hypoxic radiosensitizer and cytotoxin of superior in vitro therapeutic index to the fully aromatic nitroimidazole-linked acridine series, which strongly bind to DNA through intercalation. Extending our investigations in this area, we have synthesized and have been evaluating five more nitroimidazoles tethered to quinoline-based chromophores. Here, the in vitro and limited in vivo results of 9-[3-(2-nitro-1-imidazolyl)-propylamino]cyclopenteno[b]quinoline hydrochloride (NLCPQ-1) are presented. On a concentration basis, NLCPQ-1 was the most potent analog so far tested of the above mentioned small series as radiosensitizer or cytotoxin of hypoxic cells. It had a hypoxic selectivity of 9-12 in various cell lines, a C1.6 of approximately equal to 7 microM, and a sensitization enhancement ratio of 3.2 at 50 microM (approximately equal to 27% of the IC50(A)) in V79 cells. Its in vitro therapeutic index (defined as IC50(A)/C1.6) was 25-30 vs. 11 for 9-[3-(2-nitro-1-imidazolyl)propylamino]acridine hydrochloride. The partition coefficient in octanol/water was 0.30 +/- 0.01. The uptake factor (intracellular vs. extracellular concentration) was increased by increasing input concentration and reached 92 at 80 microM. When NLCPQ-1 was administered i.p. at 15 mg/kg, at various time-intervals before a single, 20 Gy radiation dose in Balb/c mice bearing EMT6 tumors, significant synergism was observed. The in vivo-in vitro assay was used as an endpoint, and the fractional product analysis was used to determine synergistic interactions. No toxicity was observed at doses up to 50 mg/kg NLCPQ-1 in nontumor-bearing Balb/c mice.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , DNA/drug effects , DNA/metabolism , Nitroimidazoles/pharmacology , Nitroimidazoles/pharmacokinetics , Quinolines/pharmacology , Quinolines/pharmacokinetics , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/pharmacokinetics , Animals , Antineoplastic Agents/metabolism , Cell Line , Chemical Phenomena , Chemistry, Physical , Cricetinae , Female , Humans , Mice , Mice, Inbred BALB C , Nitroimidazoles/metabolism , Quinolines/metabolism , Radiation-Sensitizing Agents/metabolism
17.
Oncol Res ; 6(9): 439-48, 1994.
Article in English | MEDLINE | ID: mdl-7703530

ABSTRACT

Electron-affinic compounds with strong DNA intercalating properties have demonstrated less than the expected radiosensitization due to restriction of their mobility along the DNA backbone and their lower extravascular diffusion in tumors. A 2-nitroimidazole linked 1,2,3,4-tetrahydroacridine derivative (THNLA-1) has been synthesized as a hypoxia-selective cytotoxin and radiosensitizer with presumably lower DNA-binding affinity due to the perturbation of the planarity in the acridine ring. THNLA-1 is a good hypoxia-selective cytotoxin with a differential toxicity of approximately equal to 11 in V79 cells, but it is approximately equal to 2 times less potent on a concentration basis than NLA-1 (the 2-nitroimidazole linked acridine analog). However, THNLA-1 is a very efficient radiosensitizer, showing a sensitization enhancement ratio (SER) of 3.04 +/- 0.05 at 100 microM at 25 degrees C, and the concentration giving an SER of 1.6(C1.6) is 19.0 +/- 0.5 microM. The therapeutic index, defined as the ratio of the clonogenic IC50 under aerobic conditions for 1-h exposure (IC50A,1h) to the C1.6 value, is 20 for THNLA-1 vs. 11 for NLA-1. THNLA-1's partition coefficient in octanol/water is 0.14 +/- 0.02. Topoisomerase I and II interaction studies with THNLA-1 showed that topoisomerase I-mediated relaxation of supercoiled DNA was inhibited at relatively high THNLA-1 concentrations (> or = 1000 microM), while topoisomerase II-mediated decatenation of kinetoplast DNA remained unaffected even in concentrations toxic in vitro under aerobic conditions. Uptake studies under aerobic conditions showed high intracellular drug concentrations, compatible with the required ones for topoisomerase I inhibition.


Subject(s)
Aminoacridines/pharmacology , DNA/drug effects , DNA/metabolism , Intercalating Agents/pharmacology , Nitroimidazoles/pharmacology , Radiation-Sensitizing Agents/pharmacology , Tacrine/analogs & derivatives , Aerobiosis , Aminoacridines/pharmacokinetics , Aminoacridines/toxicity , Animals , Cell Hypoxia , Cells, Cultured , Cricetinae , Cricetulus , DNA Topoisomerases, Type I/drug effects , DNA Topoisomerases, Type II/drug effects , DNA, Superhelical/drug effects , DNA, Superhelical/metabolism , Intercalating Agents/pharmacokinetics , Intercalating Agents/toxicity , Lung/cytology , Lung/drug effects , Lung/metabolism , Nitroimidazoles/pharmacokinetics , Nitroimidazoles/toxicity , Radiation-Sensitizing Agents/pharmacokinetics , Tacrine/pharmacokinetics , Tacrine/toxicity
18.
Oncol Res ; 5(1): 29-36, 1993.
Article in English | MEDLINE | ID: mdl-8369573

ABSTRACT

Four 2-alkysulfonyloxy-3-hydroxy-1,4-naphthoquinone derivatives, VH(1-4), have been synthesized as a novel class of bioreductive alkylating agents and tested as hypoxic cytotoxins, radiosensitizers, and chemosensitizers of V79 cells. In this series, the naphthoquinone is the bioreductive moiety while the sulfonyloxy group, as a good leaving group, could lead to alkylations of nucleophilic substrates in the cells. All VH-compounds are moderately selective cytotoxins of V79 cells with a differential aerobic-hypoxic toxicity of approximately 2, i.e. ratio of drug concentrations to give 50% killing for aerobic versus hypoxic cells. All derivatives are radiosensitizers of hypoxic V79 cells but only VH-3 can give a sensitization enhancement ratio of 2.05 at a nontoxic concentration. Finally, all VH-derivatives are good chemosensitizers of V79 cells to the aerobic toxicity of melphalan, either under hypoxic preincubation without melphalan or under aerobic coincubation with melphalan. The dose modification factors were approximately 2, at low, non-toxic concentrations.


Subject(s)
Naphthoquinones/pharmacology , Radiation-Sensitizing Agents/pharmacology , Absorption , Alkylating Agents/pharmacokinetics , Alkylating Agents/pharmacology , Animals , Cell Hypoxia/physiology , Cells, Cultured , Cricetinae , Cricetulus , Drug Synergism , Melphalan/toxicity , Naphthoquinones/pharmacokinetics , Radiation-Sensitizing Agents/pharmacokinetics
19.
Jpn J Cancer Res ; 83(8): 907-13, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1399828

ABSTRACT

Two new bioreductive compounds, 9-[3-(2-nitro-1-imidazolyl)propylamino]acridine hydrochloride (NLA-1) and 9-[2-(2-nitro-1-imidazolyl)ethylamino]acridine hydrochloride (NLA-2), which behave as hypoxic cytotoxins and radiosensitizers, have been investigated for potentiation of 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea's (CCNU) cytotoxic activity in vitro using V-79 cells. The preincubation effect as well as conditions of coadministration of CCNU with each sensitizer have been examined. In this latter case, the median-effect analysis was applied to evaluate whether the phenomenon was additive or synergistic. A clonogenic assay was used to score survival. Both bioreductive compounds, even at very low concentrations, significantly enhance the cytotoxic activity of CCNU under conditions of hypoxic preincubation. The enhancement of CCNU cytotoxicity is dependent upon preincubation time and the concentrations of both CCNU and the specific bioreductive agent. Coincubation of cells under hypoxia with CCNU and each bioreductive agent led to some potentiation, but only at lower survival levels. No chemosensitization was observed under aerobic conditions with either sensitizer.


Subject(s)
Aminoacridines/toxicity , Cell Survival/drug effects , Chelating Agents/toxicity , Lomustine/toxicity , Nitroimidazoles/toxicity , Radiation-Sensitizing Agents/toxicity , Aminoacridines/pharmacology , Animals , Cell Line , Chelating Agents/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Kinetics , Lomustine/pharmacology , Mathematics , Nitroimidazoles/pharmacology , Radiation-Sensitizing Agents/pharmacology
20.
Jpn J Cancer Res ; 83(4): 410-4, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1506276

ABSTRACT

Two new bioreductive compounds, 9-[3-(2-nitro-1-imidazolyl)propylamino]acridine hydrochloride (NLA-1) and 9-[2-(2-nitro-1-imidazolyl)ethylamino]acridine hydrochloride (NLA-2), have been prepared. They feature an acridine ring to intercalate with DNA, a 2-nitroimidazole ring as the radiosensitizing moiety and an amino functionality for increased DNA-binding and hydrophilicity. Time and concentration dependent cytotoxicity as well as radiosensitization efficacy of the two compounds under hypoxic or aerobic conditions were determined in vitro using V-79 cells and an MTT colorimetric or clonogenic assay. The isosensitization point (ISP), defined as that drug concentration which results in the same survival decrement upon exposure of hypoxic or oxygenated cells to a given radiation dose, has been determined for both compounds at 7.5 Gy and the values are significantly lower than the ISPs of 5-[3-(2-nitro-1-imidazolyl)propyl]phenanthridinium bromide, 2-(2-nitro-1-imidazolyl)ethylamine or misonidazole (MISO). NLA-1 and NLA-2 are potent hypoxic cytotoxins and on a concentration basis, more potent than MISO as radiosensitizers in vitro. The sensitization enhancement ratios were significantly increased when 1 h drug preincubation under hypoxia at 37 degrees C was applied, before irradiation at room temperature.


Subject(s)
Aminoacridines/pharmacology , Cell Survival/drug effects , Nitroimidazoles/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Cell Hypoxia , Cell Line , Kinetics , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...