Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Cancer Chemother Pharmacol ; 85(3): 573-583, 2020 03.
Article in English | MEDLINE | ID: mdl-31915968

ABSTRACT

Treatment with fludarabine phosphate (9-ß-D-arabinofuranosyl-2-F-adenine 5'-phosphate, F-araAMP) leads to regressions and cures of human tumor xenografts that express Escherichia coli purine nucleoside phosphorylase (EcPNP). This occurs despite the fact that fludarabine (F-araA) is a relatively poor substrate for EcPNP, and is cleaved to liberate 2-fluoroadenine at a rate only 0.3% that of the natural E. coli PNP substrate, adenosine. In this study, we investigated a panel of naturally occurring PNPs to identify more efficient enzymes that may be suitable for metabolizing F-araA as part of experimental cancer therapy. We show that Trichomonas vaginalis PNP (TvPNP) cleaves F-araA with a catalytic efficiency 25-fold greater than the prototypic E. coli enzyme. Cellular extracts from human glioma cells (D54) transduced with lentivirus stably expressing TvPNP (D54/TvPNP) were found to cleave F-araA at a rate similar to extracts from D54 cells expressing EcPNP, although much less enzyme was expressed per cell in the TvPNP transduced condition. As a test of safety and efficacy using TvPNP, human head and neck squamous cell carcinoma (FaDu) xenografts expressing TvPNP were studied in nude mice and shown to exhibit robust tumor regressions, albeit with partial weight loss that resolved post-therapy. F-araAMP was also a very effective treatment for mice bearing D54/TvPNP xenografts in which approximately 10% of tumor cells expressed the enzyme, indicating pronounced ability to kill non-transduced tumor cells (high bystander activity). Moreover, F-araAMP demonstrated activity against D54 tumors injected with an E1, E3 deleted adenoviral vector encoding TvPNP. In that setting, despite higher F-araA cleavage activity using TvPNP, tumor responses were similar to those obtained with EcPNP, indicating factors other than F-Ade production may limit regressions of the D54 murine xenograft model. Our results establish that TvPNP is a favorable enzyme for activating F-araA, and support further studies in combination with F-araAMP for difficult-to-treat human cancers.


Subject(s)
Glioma/drug therapy , Purine-Nucleoside Phosphorylase/genetics , Trichomonas vaginalis/enzymology , Vidarabine/analogs & derivatives , Animals , Cell Line, Tumor , Escherichia coli/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Glioma/genetics , Humans , Lentivirus/genetics , Mice , Mice, Nude , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Vidarabine/pharmacology
2.
Article in English | MEDLINE | ID: mdl-23004929

ABSTRACT

Thiarabine was evaluated for antitumor activity in seven human leukemia, lymphoma, and myeloma xenograft models to explore the activity in hematological malignancies. Thiarabine was active against all of the human leukemia and lymphoma lines tested, being curative against HL-60 leukemia and AS283 lymphoma and effecting tumor regressions in CCRF-CEM, MOLT-4, and K-562 leukemia and RL lymphoma models, but did not exhibit any appreciable activity against RPMI-8226 myeloma. For the leukemia/lymphoma models, thiarabine was more efficacious than ara-C/palmO-ara-C (four models), clofarabine (three models), fludarabine monophosphate (five models), cladribine (four models), and gemcitabine (six models). Thiarabine warrants future clinical trials with leukemias/lymphomas.


Subject(s)
Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Leukemia/drug therapy , Lymphoma/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Arabinonucleosides/administration & dosage , Cell Line, Tumor , Drug Evaluation, Preclinical , HL-60 Cells , Humans , K562 Cells , Mice , Treatment Outcome
3.
Article in English | MEDLINE | ID: mdl-23004932

ABSTRACT

Clofarabine, an approved anticancer drug, was evaluated in combination with radiation in six subcutaneously implanted human tumor xenograft models. Clofarabine had no effect on the growth of SF-295 glioblastoma, which was not enhanced by radiation. There was no difference between clofarabine with and without radiation in the DU-145 prostate model. The combined effect on NCI-H460 lung tumors appeared to be additive. SR475 head and neck, PANC-1 pancreatic, and HCT-116 colon tumors were radiomodified by clofarabine. The radiomodifying capacity of clofarabine was superior to that for gemcitabine in two models (PANC-1 and HCT-116) and was comparable in the other four models.


Subject(s)
Adenine Nucleotides/therapeutic use , Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Neoplasms, Experimental/therapy , Adenine Nucleotides/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Arabinonucleosides/administration & dosage , Cell Line, Tumor , Clofarabine , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Drug Evaluation, Preclinical , Humans , Mice , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/radiotherapy , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
4.
Article in English | MEDLINE | ID: mdl-22908953

ABSTRACT

Thiarabine is undergoing clinical trials. In support of that effort combination therapy of thiarabine plus six clinical anticancer agents was evaluated using various human tumor xenograft models. The antitumor activity of thiarabine in combination appeared to be greater than additive with irinotecan (DLD-1 colon), paclitaxel (PC-3 prostate), cisplatin (PC-3 prostate), or cyclophosphamide (RL lymphoma), additive with irinotecan (NCI-H460 NSCLC), cisplatin (NCI-H460 NSCLC) or methotrexate (CCRF-CEM leukemia), and less than additive with irinotecan (HT29 colon), paclitaxel (NCI-H460 NSCLC) or cisplatin (NCI-H23 NSCLC). Combining thiarabine with irinotecan, paclitaxel, cisplatin, or cyclophosphamide should receive consideration in the clinical treatment of cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arabinonucleosides/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Arabinonucleosides/administration & dosage , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cell Line, Tumor , Cisplatin/administration & dosage , Cyclophosphamide/administration & dosage , Female , HT29 Cells , Humans , Irinotecan , Male , Mice , Mice, Nude , Mice, SCID , Neoplasms/pathology , Paclitaxel/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
Cancer Chemother Pharmacol ; 70(2): 321-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22760227

ABSTRACT

PURPOSE: Systemically administered fludarabine phosphate (F-araAMP) slows growth of human tumor xenografts that express Escherichia coli purine nucleoside phosphorylase (PNP). However, this treatment has been limited by the amount of F-araAMP that can be administered in vivo. The current study was designed to (1) determine whether efficacy of this overall strategy could be improved by intratumoral administration of F-araAMP, (2) test enhancement of the approach with external beam radiation, and (3) optimize recombinant adenovirus as a means to augment PNP delivery and bystander killing in vivo. METHODS: The effects of systemic or intratumoral F-araAMP in mice were investigated with human tumor xenografts (300 mg), in which 10 % of the cells expressed E. coli PNP from a lentiviral promoter. Tumors injected with an adenoviral vector expressing E. coli PNP (Ad/PNP; 2 × 10(11) viral particles, 2 times per day × 3 days) and the impact of radiotherapy on tumors treated by this approach were also studied. Radiolabeled F-araAMP was used to monitor prodrug activation in vivo. RESULTS: Intratumoral administration of F-araAMP in human tumor xenografts expressing E. coli PNP resulted in complete regressions and/or prolonged tumor inhibition. External beam radiation significantly augmented this effect. Injection of large human tumor xenografts (human glioma, nonsmall cell lung cancer, or malignant prostate tumors) with Ad/PNP followed by intratumoral F-araAMP resulted in excellent antitumor activity superior to that observed following systemic administration of prodrug. CONCLUSION: Activation of F-araAMP by E. coli PNP results in destruction of large tumor xenografts in vivo, augments radiotherapy, and promotes robust bystander killing. Our results indicate that intratumoral injection of F-araAMP leads to ablation of tumors in vivo with minimal toxicity.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Drug Resistance, Neoplasm/drug effects , Genetic Therapy , Prodrugs/therapeutic use , Purine-Nucleoside Phosphorylase/genetics , Vidarabine Phosphate/analogs & derivatives , Adenoviridae/genetics , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Bystander Effect/drug effects , Bystander Effect/genetics , Bystander Effect/radiation effects , Cell Line, Tumor , Combined Modality Therapy , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/radiation effects , Escherichia coli/genetics , Genetic Vectors , Glioma/drug therapy , Glioma/genetics , Glioma/radiotherapy , Humans , Injections, Intralesional , Mice , Mice, Nude , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Purine-Nucleoside Phosphorylase/metabolism , Transfection , Transplantation, Heterologous , Vidarabine Phosphate/administration & dosage , Vidarabine Phosphate/pharmacokinetics , Vidarabine Phosphate/therapeutic use
6.
Article in English | MEDLINE | ID: mdl-22257207

ABSTRACT

A murine P388 leukemia line fully resistant to thiarabine was obtained after five courses of intraperitoneal treatment (daily for nine consecutive days). The subline was sensitive as was the parental P388/0 line to 5-fluorouracil, gemcitabine, cyclophosphamide, cisplatin, melphalan, BCNU, mitomycin C, doxorubicin, mitoxantrone, etoposide, irinotecan, vincristine, and paclitaxel, but was cross resistant (at least marginally) to three antimetabolites: palmO-ara-C, fludarabine phosphate, and methotrexate. The deoxycytidine kinase activity in the subline was comparable to that for P388/0, whereas the dCMP deaminase activity was 43% of that for P388/0. No deoxycytidine deaminase activity was detected in either of the leukemias. There appeared to be little, if any, difference in the metabolism of deoxycytidine, cytidine, or thiarabine in the two leukemias.


Subject(s)
Antimetabolites/administration & dosage , Antineoplastic Agents/administration & dosage , Arabinonucleotides/administration & dosage , Cell Line, Tumor/drug effects , Drug Resistance, Neoplasm/drug effects , Animals , Antimetabolites/chemical synthesis , Antineoplastic Agents/chemistry , Arabinonucleotides/chemical synthesis , Cell Line, Tumor/cytology , Cell Line, Tumor/enzymology , DCMP Deaminase/metabolism , Deoxycytidine Kinase/metabolism , Female , Leukemia P388 , Mice , Neoplasm Transplantation , Transplantation, Heterologous
7.
Eur J Med Chem ; 47(1): 167-74, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22112758

ABSTRACT

A series of C-6 alkyl, cycloalkyl, and aryl-9-(ß-d-ribofuranosyl)purines were synthesized and their substrate activities with Escherichia coli purine nucleoside phosphorylase (E. coli PNP) were evaluated. (Ph(3)P)(4)Pd-mediated cross-coupling reactions of 6-chloro-9-(2,3,5-tri-O-acetyl-ß-d-ribofuranosyl)-purine (6) with primary alkyl (Me, Et, n-Pr, n-Bu, isoBu) zinc halides followed by treatment with NH(3)/MeOH gave the corresponding 6-alkyl-9-(ß-d-ribofuranosyl)purine derivatives 7-11, respectively, in good yields. Reactions of 6 with cycloalkyl(propyl, butyl, pentyl)zinc halides and aryl (phenyl, 2-thienyl)zinc halides gave under similar conditions the corresponding 6-cyclopropyl, cyclobutyl, cyclopentyl, phenyl, and thienyl -9-(ß-d-ribofuranosyl)purine derivatives 12-16, respectively in high yields. E. coli PNP showed a high tolerance to the steric and hydrophobic environment at the 6-position of the synthesized purine ribonucleosides. Significant cytotoxic activity was observed for 8, 12, 15, and 16. Evaluation of 12 and 16 against human tumor xenografts in mice did not demonstrate any selective antitumor activity. In addition, 6-methyl-9-(ß-d-arabinofuranosyl)purine (18) was prepared and evaluated.


Subject(s)
Escherichia coli/enzymology , Halogenation , Palladium/chemistry , Purine Nucleosides/chemistry , Purine Nucleosides/metabolism , Purine-Nucleoside Phosphorylase/metabolism , Ribonucleosides/chemistry , Ribonucleosides/metabolism , Zinc/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Catalysis , Cell Line , Cell Line, Tumor , Humans , Mice , Purine Nucleosides/chemical synthesis , Purine Nucleosides/pharmacology , Ribonucleosides/chemical synthesis , Ribonucleosides/pharmacology , Xenograft Model Antitumor Assays
8.
PLoS One ; 6(12): e29290, 2011.
Article in English | MEDLINE | ID: mdl-22195040

ABSTRACT

The Wnt/ß-catenin signaling pathway is important for tumor initiation and progression. The low density lipoprotein receptor-related protein-6 (LRP6) is an essential Wnt co-receptor for Wnt/ß-catenin signaling and represents a promising anticancer target. Recently, the antihelminthic drug, niclosamide was found to inhibit Wnt/ß-catenin signaling, although the mechanism was not well defined. We found that niclosamide was able to suppress LRP6 expression and phosphorylation, block Wnt3A-induced ß-catenin accumulation, and inhibit Wnt/ß-catenin signaling in HEK293 cells. Furthermore, the inhibitory effects of niclosamide on LRP6 expression/phosphorylation and Wnt/ß-catenin signaling were conformed in human prostate PC-3 and DU145 and breast MDA-MB-231 and T-47D cancer cells. Moreover, we showed that the mechanism by which niclosamide suppressed LRP6 resulted from increased degradation as evident by a shorter half-life. Finally, we demonstrated that niclosamide was able to induce cancer cell apoptosis, and displayed excellent anticancer activity with IC(50) values less than 1 µM for prostate PC-3 and DU145 and breast MDA-MB-231 and T-47D cancer cells. The IC(50) values are comparable to those shown to suppress the activities of Wnt/ß-catenin signaling in prostate and breast cancer cells. Our data indicate that niclosamide is a unique small molecule Wnt/ß-catenin signaling inhibitor targeting the Wnt co-receptor LRP6 on the cell surface, and that niclosamide has a potential to be developed a novel chemopreventive or therapeutic agent for human prostate and breast cancer.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Neoplasms/pathology , Niclosamide/pharmacology , Proteolysis/drug effects , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dishevelled Proteins , Drug Screening Assays, Antitumor , Female , HEK293 Cells , Humans , Male , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Prostatic Neoplasms/pathology , Wnt3A Protein/pharmacology
9.
Nucleosides Nucleotides Nucleic Acids ; 30(11): 826-38, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22060549

ABSTRACT

A murine P388 leukemia line fully resistant to clofarabine was obtained after only two courses of intraperitoneal treatment (three times a day for nine consecutive days). The resistance was stable for at least 13 weeks without treatment. The subline was as sensitive to 5-fluorouracil, methotrexate, cyclophosphamide, cisplatin, melphalan, BCNU, doxorubicin, etoposide, irinotecan, vincristine, and docetaxel as was the parental P388/0 line but was cross-resistant to five antimetabolites [palmO-ara-C, 4'-thio-ara-C, fludarabine phosphate, cladribine, and gemcitabine-all of which require deoxycytidine kinase for activation] and paclitaxel. The subline had less than 1% of the deoxycytidine kinase activity in comparison to P388/0.


Subject(s)
Adenine Nucleotides/therapeutic use , Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Cell Line, Tumor/drug effects , Drug Resistance, Neoplasm , Leukemia P388/drug therapy , Adenine Nucleotides/pharmacology , Animals , Antineoplastic Agents/pharmacology , Arabinonucleosides/pharmacology , Cell Line, Tumor/enzymology , Cell Separation , Clofarabine , Deoxycytidine Kinase/metabolism , Leukemia P388/enzymology , Mice
10.
FEBS Lett ; 585(19): 3120-5, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21907199

ABSTRACT

Mesd is a specialized chaperone for Wnt co-receptor low-density lipoprotein receptor-related protein-5 (LRP5) and LRP6, which contain four ß-propeller/epidermal growth factor modules, named E1 to E4 from N- to C-terminal, in their extracellular domains. Herein, we demonstrated that recombinant Mesd protein is a general Wnt inhibitor that blocks Wnt/ß-catenin signaling induced not only by LRP6 E1-E2-binding Wnts but also by LRP6 E3-E4-binding Wnts. We also found that Mesd suppressed Wnt/ß-catenin signaling induced by Wnt1 in prostate cancer PC-3 cells, and inhibited tumor growth in PC-3 xenograft model. Our results indicate that Mesd is a universal inhibitor of Wnt/LRP signaling on the cell surface.


Subject(s)
Ligands , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Molecular Chaperones/metabolism , Signal Transduction/physiology , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Male , Mice , Molecular Chaperones/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Transplantation, Heterologous , Wnt Proteins/genetics , beta Catenin/metabolism
11.
Cancer Biol Ther ; 11(7): 627-32, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21301207

ABSTRACT

Many solid tumors and hematologic malignancies lack expression of the enzyme methylthioadenosine phosphorylase (MTAP), due either to deletion of the MTAP gene or to methylation of the MTAP promoter. In cells that have MTAP, its natural substrate, methylthioadenosine (MTA), generated during polyamine biosynthesis, is cleaved to adenine and 5-methylthioribose-1-phosphate. The latter compound is further metabolized to methionine. Adenine and methionine are further metabolized and hence salvaged. In MTAP-deficient cells, however, MTA is not cleaved and the salvage pathway for adenine and methionine is absent. As a result, MTAP-deficient cells are more sensitive than MTAP-positive cells to inhibitors of de novo purine synthesis and to methionine deprivation. The challenge has been to take advantage of MTAP deficiency, and the changes in metabolism that follow, to design a strategy for targeted treatment. In this review, the frequency of MTAP-deficiency is presented and past and recent strategies to target such deficient cells are discussed, including one in which MTA is administered, followed by very high doses of a toxic purine or pyrimidine analog. In normal host cells, adenine, generated from MTA, blocks conversion of the analog to its toxic nucleotide. In MTAP-deficient tumor cells, conversion proceeds and the tumor cells are selectively killed. Successful mouse studies using this novel strategy were recently reported.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/enzymology , Purine-Nucleoside Phosphorylase/deficiency , Animals , Humans , Molecular Targeted Therapy , Purines/antagonists & inhibitors , Purines/biosynthesis
12.
Cancer Chemother Pharmacol ; 67(4): 855-65, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20567829

ABSTRACT

UNLABELLED: Hypoxia is a common feature of solid tumors. Up-regulation of hypoxia-inducing factor-1 (HIF-1) occurs in the majority of primary malignant tumors and in two-thirds of metastases, while most normal tissues are negative. HIF-1 induces the glycolytic phenotype, which creates an acidic extracellular microenvironment and associated pH gradient such that drugs that are weak acids are selectively taken up and retained in acidic tumors. 7-Butyl-10-amino-camptothecin (BACPT) is a prime example of an agent that can exploit the tumor pH gradient for enhanced selectivity. PURPOSE: This study profiles the antitumor activity of BACPT in vitro and its water-soluble dipeptide ester, BACPTDP, in vivo. METHODS: Antitumor activity was evaluated by proliferation assays in cancer cell lines and in murine xenograft models for human neuroblastoma (IMR-32), colon (HT29), ovarian (SK-OV-3), pancreatic (Panc-1), glioma (SF-295) and non-small-cell lung (NCI-H460) cancers. RESULTS: BACPT had superior antiproliferative activity compared to established drugs in monolayer cultures of human neuroblastoma and pancreatic tumor cell lines and in 3-dimensional histocultures of colon and primary ovarian cancer. Antitumor activity of BACPTDP was comparable to irinotecan in IMR-32, HT29, SF-295 and NCI-H460 xenografts, significantly greater in SK-OV-3 and in Panc-1 where complete regressions were observed. Combination of BACPT with gemcitabine produced additive to synergistic interactions in Panc-1 cells that were independent of drug ratio and optimal when gemcitabine was administered 24 h prior to BACPT. CONCLUSIONS: BACPTDP is a water-soluble camptothecin pro-drug that spontaneously generates the lipid-soluble active agent, BACPT. This topoisomerase inhibitor exploits solid tumor physiology for improved selectivity and activity against multiple tumor types with particular promise for use in treating pediatric neuroblastoma and pancreatic carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Dipeptides/pharmacology , Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Camptothecin/administration & dosage , Camptothecin/pharmacology , Cell Hypoxia , Cell Proliferation/drug effects , Dipeptides/administration & dosage , Drug Administration Schedule , Drug Synergism , Female , Humans , Hydrogen-Ion Concentration , Mice , Mice, Nude , Neoplasms/pathology , Prodrugs , Xenograft Model Antitumor Assays
13.
Cancer Chemother Pharmacol ; 68(2): 399-403, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21069340

ABSTRACT

PURPOSE: 4'-Thio-ß-D-arabinofuranosylcytosine (4'-thio-ara-C), which has shown a broad spectrum of antitumor activity against human tumor systems in mice and is undergoing clinical trials, was evaluated for cross-resistance to seven clinical agents in order to identify potentially useful guides for patient selection for further clinical trials of 4'-thio-ara-C and possible noncross-resistant drug combinations with 4'-thio-ara-C. METHODS: A drug resistance profile for 4'-thio-ara-C, which was administered intraperitoneally daily for nine consecutive days, was obtained using seven drug-resistant P388 and L1210 leukemias that were implanted intraperitoneally in mice. RESULTS: Multidrug-resistant P388 leukemias (leukemias resistant to doxorubicin, etoposide, or paclitaxel) exhibited no cross-resistance to 4'-thio-ara-C. Leukemias resistant to camptothecin, cisplatin, and 5-fluorouracil were also not cross-resistant to 4'-thio-ara-C. Only the leukemia resistant to 1-ß-D-arabinofuranosylcytosine was cross-resistant to 4'-thio-ara-C. CONCLUSIONS: The data suggest that (1) it may be important to exclude or to monitor with extra care patients who have previously been treated with 1-ß-D-arabinofuranosylcytosine and (2) the lack of cross-resistance seen with 4'-thio-ara-C may contribute to therapeutic synergism when 4'-thio-ara-C is combined with other agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Leukemia L1210/drug therapy , Leukemia P388/drug therapy , Algorithms , Animals , Clinical Trials as Topic , Humans , Leukemia/drug therapy , Mice , Mice, Inbred Strains , Survival Analysis
14.
Cancer Chemother Pharmacol ; 64(4): 829-35, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19255760

ABSTRACT

PURPOSE: The purpose of this investigation was to synthesize a series of carbonate and carbamate derivatives of 4-demethylpenclomedine (DM-PEN), the major plasma non-toxic metabolite of penclomedine (PEN) seen in patients. DM-PEN has been observed to be an active antitumor agent in mouse human xenograft tumor models and non-neurotoxic in a rat model, however, activity in intracranially implanted human glioma xenograft models have not been reported. The major goal was to identify derivatives that are active in brain tumors. METHODS: Derivatives were prepared from DM-PEN and evaluated in vivo against human U251 glioblastoma, D54 glioblastoma and MX-1 breast tumor xenografts and mammary tumor 16/C that were implanted in the mammary fat pad or intracranially (IC). RESULTS: Carbonate and carbamate derivatives were found to be superior to DM-PEN against IC growing human glioblastoma xenografts. CONCLUSION: The activity of the carbonates and carbamates against human tumor xenografts in vivo suggests consideration of these two series of derivatives of DM-PEN for clinical development.


Subject(s)
Antineoplastic Agents/therapeutic use , Carbamates/chemistry , Carbonates/chemistry , Picolines/therapeutic use , Animals , Antineoplastic Agents/chemistry , Chromatography, Thin Layer , Humans , Magnetic Resonance Spectroscopy , Mice , Picolines/chemistry , Rats , Spectrometry, Mass, Fast Atom Bombardment , Spectrophotometry, Ultraviolet , Transplantation, Heterologous
15.
Cancer Chemother Pharmacol ; 64(2): 253-61, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19002461

ABSTRACT

PURPOSE: Clofarabine increases the activation of 1-beta-D-arabinofuranosyl cytosine (araC) in tumor cells, and combination of these two drugs has been shown to result in good clinical activity against various hematologic malignancies. 1-beta-D-[4-thio-arabinofuranosyl] cytosine (T-araC) is a new cytosine analog that has exhibited excellent activity against a broad spectrum of human solid tumors and leukemia/lymphoma xenografts in mice and is currently being evaluated in patients as a new drug for the treatment of cancer. Since T-araC has a vastly superior preclinical efficacy profile in comparison to araC, we have initiated studies to determine the potential value of clofarabine/T-araC combination therapy. METHODS: In vitro studies have been conducted to determine the effect of clofarabine on the metabolism of T-araC, and in vivo studies have been conducted to determine the effect of the clofarabine/T-araC combination on five human tumor xenografts in mice. RESULTS: Initial studies with various tumor cells in culture indicated that a 2-h incubation with clofarabine enhanced the metabolism of T-araC 24 h after its removal by threefold in three tumor cell types (HCT-116 colon, K562 leukemia, and RL lymphoma) and by 1.5-fold in two other tumor cell types (MDA-MB-435 breast (melanoma), and HL-60 leukemia). Pretreatment with clofarabine resulted in a slight decrease in metabolism of T-araC in RPMI-8226 myeloma cells (65% of control) and inhibited metabolism of T-araC in CCRF-CEM leukemia cells by 90%. In vivo combination studies were conducted with various human tumor xenografts to determine whether or not the modulations observed in vitro were reflective of the in vivo situation. Clofarabine and T-araC were administered on alternate days for five treatments each (q2dx5) with the administration of T-araC 24 h after each clofarabine treatment. Combination treatment of HCT-116, K562, HL-60, or RL tumors with clofarabine and T-araC resulted in dramatically superior anti-tumor activity than treatment with either agent alone, whereas this combination resulted in antagonism in CCRF-CEM tumors. The in vivo antitumor activity of clofarabine plus T-araC against HCT-116 tumors was much better than the activity seen with clofarabine plus araC. CONCLUSIONS: These studies provide a rationale for clinical trials using this combination in the treatment of acute leukemias as well as solid tumors and suggest that this combination would exhibit greater antitumor activity than that of clofarabine plus araC.


Subject(s)
Adenine Nucleotides/therapeutic use , Antineoplastic Agents/pharmacology , Arabinonucleosides/therapeutic use , Neoplasms, Experimental/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Clofarabine , Drug Synergism , Drug Therapy, Combination , Humans , Mice , Mice, Nude , Mice, SCID , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Nucleosides Nucleotides Nucleic Acids ; 28(5): 657-77, 2009 May.
Article in English | MEDLINE | ID: mdl-20183608

ABSTRACT

As part of an ongoing program to develop novel antitumor agents over the years, we have synthesized and evaluated a number of 4'-C-substituted nucleosides. A few years ago, we reported the first synthesis of 4'-C-hydroxymethyl-2'-fluoro arabino nucleosides, which did not exhibit any cytotoxicity. In our exploration of related compounds, we synthesized and evaluated the 4'-C-methyl-2'-fluoro arabino nucleosides in both the purine and pyrimidine series. In the pyrimidine series, 1-(4-C-methyl-2-fluoro-beta-D-arabinofuranosyl) cytosine (13) was found to be highly cytotoxic and had significant antitumor activity in mice implanted with human tumor xenografts. The synthesis and anticancer activity of this series of nucleosides are reported.


Subject(s)
Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Neoplasms/drug therapy , Pyrimidine Nucleosides/therapeutic use , Pyrimidine Nucleotides/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Arabinonucleosides/chemical synthesis , Arabinonucleosides/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Halogenation , Humans , Mice , Pyrimidine Nucleosides/chemical synthesis , Pyrimidine Nucleosides/chemistry , Pyrimidine Nucleotides/chemical synthesis , Pyrimidine Nucleotides/chemistry
17.
Int J Radiat Oncol Biol Phys ; 70(1): 213-20, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18037589

ABSTRACT

PURPOSE: Combination treatment with radiotherapy and chemotherapy has emerged as the dominant form of cancer adjuvant regimens in recent years. Clofarabine, a newly approved drug for pediatric leukemia, is a second-generation purine nucleoside analogue that can block DNA synthesis and inhibit DNA repair. Therefore, we hypothesized that clofarabine could work synergistically with radiotherapy to increase the tumor cell response. METHODS AND MATERIALS: The effects of clofarabine on radiosensitivity have been established in several tumor cell lines in vitro and in vivo using colony-forming assays and tumor xenografts. The effect of clofarabine on the DNA damage response was also studied in vitro by measuring gamma-H2AX focus formation. RESULTS: Clonogenic survival was significantly reduced in irradiated cells treated with clofarabine, demonstrating the strong radiosensitizing effect of clofarabine. Furthermore, clofarabine displayed a radiosensitizing effect that was greater than gemcitabine or 5-fluorouracil. We also found that low doses of clofarabine can prolong the presence of radiation-induced gamma-H2AX nuclear focus formation, and high doses of clofarabine can induce DNA double-strand breaks, suggesting that clofarabine can interfere with DNA damage response pathways. In addition, clofarabine-induced radiosensitization was also established in vivo using a colorectal cancer model, DLD-1, in athymic nude mice. When combined with fractionated radiotherapy, a moderate dose of clofarabine led to a significant increase in tumor growth inhibition. CONCLUSION: Clofarabine acts as a powerful radiosensitizer both in vitro and in vivo by interfering with the DNA damage response.


Subject(s)
Adenine Nucleotides/pharmacology , Arabinonucleosides/pharmacology , DNA Damage , DNA Repair/drug effects , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Animals , Cell Line, Tumor , Clofarabine , DNA, Neoplasm/drug effects , DNA, Neoplasm/radiation effects , HeLa Cells , Histones/metabolism , Humans , Male , Mice , Mice, Nude , Radiation Dosage , Xenograft Model Antitumor Assays
18.
J Chem Inf Model ; 47(5): 1897-905, 2007.
Article in English | MEDLINE | ID: mdl-17676832

ABSTRACT

In silico chemical library screening (virtual screening) was used to identify a novel lead compound capable of inhibiting S-adenosylmethionine decarboxylase (AdoMetDC). AdoMetDC is intimately involved in the biosynthesis of polyamines, which are essential for tumor progression and are elevated in numerous types of tumors. Therefore, inhibition of this enzyme provides an attractive target for the discovery of novel anticancer drugs. We performed virtual screening using a computer model derived from the X-ray crystal structure of human AdoMetDC and the National Cancer Institute's Diversity Set (1990 compounds). Our docking study suggested several compounds that could serve as drug candidates since their docking modes and scores revealed potential inhibitory activity toward AdoMetDC. Experimental testing of the top-scoring compounds indicated that one of these compounds (NSC 354961) possesses an IC50 in the low micromolar range. A search of the entire NCI compound collection for compounds similar to NSC 354961 yielded two additional compounds that exhibited activity in the experimental assay but with significantly diminished potency relative to NSC 354961. In this report, we disclose the activity of NSC 354961 against AdoMetDC and its probable binding mode based on computational modeling. We also discuss the importance of virtual screening in the context of enzymes that are not readily amenable to high-throughput assays, thereby demonstrating the efficacy of virtual screening, combined with selective experimental testing, in identifying new potential drug candidates.


Subject(s)
Adenosylmethionine Decarboxylase/antagonists & inhibitors , Aminacrine/chemistry , Aminacrine/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Aminacrine/chemical synthesis , Carbon Dioxide/analysis , Carbon Dioxide/chemistry , Computer Simulation , Computers , Crystallography, X-Ray , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Humans , Recombinant Proteins/chemistry , Reproducibility of Results , Software , Structure-Activity Relationship
19.
J Med Chem ; 50(14): 3283-9, 2007 Jul 12.
Article in English | MEDLINE | ID: mdl-17569517

ABSTRACT

Two boron-containing, ortho-icosahedral carborane lipophilic antifolates were synthesized, and the crystal structures of their ternary complexes with human dihydrofolate reductase (DHFR) and dihydronicotinamide adenine dinucleotide phosphate were determined. The compounds were screened for activity against DHFR from six sources (human, rat liver, Pneumocystis carinii, Toxoplasma gondii, Mycobacterium avium, and Lactobacillus casei) and showed good to modest activity against these enzymes. The compounds were also tested for antibacterial activity against L. casei, M. tuberculosis H37Ra, and three M. avium strains and for cytotoxic activity against seven different human tumor cell lines. Antibacterial and cytotoxic activity was modest, with one sample, the closo-carborane 4, showing about 10-fold greater activity. The less toxic nido-carborane 2 was also tested as a candidate for boron neutron capture therapy, but showed poor tumor retention and low selectivity ratios for boron distribution in tumor tissue versus normal tissue.


Subject(s)
Boron/chemistry , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/pharmacology , Animals , Boron Neutron Capture Therapy , Cell Line, Tumor , Crystallography, X-Ray , Folic Acid Antagonists/chemistry , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Rats , Tetrahydrofolate Dehydrogenase/drug effects
20.
Mol Cancer Ther ; 6(2): 496-505, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17308048

ABSTRACT

A major limitation of adenovirus type 5-mediated cancer gene therapy is the inefficient infection of many cancer cells. Previously, we showed that treatment with low doses of the histone deacetylase inhibitor FK228 (FR901228, depsipeptide) increased coxsackie adenovirus receptor (CAR) levels, histone H3 acetylation, and adenovirus infection efficiencies as measured by viral transgene expression in cancer cell lines but not in cultured normal cells. To evaluate FK228 in vivo, the effects of FK228 therapy in athymic mice bearing LOX IMVI or UACC-62 human melanoma xenografts were examined. Groups of mice were treated with FK228 using several dosing schedules and the differences between treated and control animals were determined. In mice with LOX IMVI xenografts (n = 6), maximum CAR induction was observed 24 h following a single FK228 dose of 3.6 mg/kg with a 13.6 +/- 4.3-fold (mean +/- SD) increase in human CAR mRNA as determined by semiquantitative reverse transcription-PCR analysis. By comparison, mouse CAR levels in liver, kidney, and lung from the same animals showed little to no change. Maximum CAR protein induction of 9.2 +/- 4.8-fold was achieved with these treatment conditions and was associated with increased histone H3 acetylation. Adenovirus carrying a green fluorescent protein (GFP) transgene (2 x 10(9) viral particles) was injected into the xenografts and GFP mRNA levels were determined. A 7.4 +/- 5.2-fold increase in GFP mRNA was found 24 h following adenovirus injection into optimally FK228-treated mice (n = 10). A 4-fold increase in GFP protein-positive cells was found following FK228 treatment. These studies suggest that FK228 treatment prior to adenovirus infection could increase the efficiency of adenovirus gene therapy in xenograft model systems.


Subject(s)
Adenoviruses, Human/genetics , Antibiotics, Antineoplastic/pharmacology , Depsipeptides/pharmacology , Histone Deacetylase Inhibitors , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Transgenes/drug effects , Acetylation , Adenoviridae Infections/drug therapy , Adenoviridae Infections/metabolism , Adenoviridae Infections/virology , Animals , Blotting, Western , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histones/metabolism , Humans , Melanoma/metabolism , Melanoma/virology , Mice , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/metabolism , Skin Neoplasms/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...