Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Ovarian Res ; 8: 62, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26384969

ABSTRACT

BACKGROUND: Ovarian cancer is the most fatal of gynaecological malignancies, usually detected at a late stage with intraperitoneal dissemination. Appropriate preclinical models are needed that recapitulate both the histopathological and molecular features of human ovarian cancer for drug-efficacy analysis. METHODS: Longitudinal studies comparing cisplatin performance either alone or in a novel cisplatin-based delivery-system, cucurbit[7]uril-encapsulated cisplatin (cisplatin@CB[7]) were performed on subcutaneous (s.c.) and intraperitoneal (i.p.) xenografts using the human ovarian cancer cell line A2780 stably expressing the small GTPase Rab25, which allows A2780 intraperitoneal growth; and luciferase, to allow tumour load measurement by non-invasive bioluminescent imaging. RESULTS: Rab25 expression induced cisplatin resistance compared to the parental cell line as assessed by the MTT assay in vitro. These findings did not translate in vivo, where cisplatin resistance was determined by the microenvironment. Subcutaneous xenografts of either parental A2780 or cisplatin-resistant Rab25-expressing A2780 cells presented similar responses to cisplatin treatment. In contrast, increased cisplatin resistance was only detected in i.p. tumours. Treatment of the cisplatin-resistant i.p. model with the novel cisplatin@CB[7] delivery system resulted in a substantial reduction of i.p. tumour load and increased necrosis. CONCLUSIONS: Poor clinical performance of novel chemotherapeutics might reflect inappropriate preclinical models. Here we present an ovarian i.p. model that recapitulates the histopathological and chemoresistant features of the clinical disease. In addition, we demonstrate that the novel cisplatin-delivery system, cisplatin@CB[7] may have utility in the treatment of drug-resistant ovarian human cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Ovarian Neoplasms/drug therapy , Proteins/metabolism , Animals , Antineoplastic Agents/administration & dosage , Capsules , Carcinoma/drug therapy , Cell Line, Tumor , Cisplatin/administration & dosage , Drug Resistance, Neoplasm , Female , Heterografts/metabolism , Injections , Mice, Nude , Neoplasm Transplantation , Peritoneal Neoplasms/drug therapy , Transplantation, Heterologous , Tumor Suppressor Protein p53/metabolism
2.
J Inorg Biochem ; 134: 100-5, 2014 May.
Article in English | MEDLINE | ID: mdl-24595010

ABSTRACT

The anticancer drug cisplatin was encapsulated within the cucurbit[7]uril macrocycle to form the host-guest complex: cisplatin@CB[7]. This was then incorporated into gelatin and 0-4% w/v polyvinyl alcohol (PVA)-based hydrogels as slow release drug delivery vehicles. The hydrogels demonstrated predicable swelling and disintegration dependent on the PVA concentration. The hydrogel with the highest PVA content was slower to swell and release drug compared with lower concentrations of PVA. The effect of the hydrogel PVA concentration on in vitro cytotoxicity was examined using A2780/CP70 ovarian cancer cells. Over the 24h drug exposure time used, hydrogels containing 4% PVA showed a 20% decrease in viable cells compared to the control, whereas hydrogels containing 0% and 2% PVA induced an 80% and 45% inhibition of cell growth, respectively. There was no measurable difference in the in vitro cytotoxicity of free cisplatin and cisplatin@CB[7] containing hydrogels. Finally, the in vivo effectiveness of a 2%-PVA hydrogel implanted under the skin of nude mice bearing A2780/CP70 xenografts showed that low dose hydrogels containing cisplatin@CB[7] (30 µg equivalent of drug) was just as effective as an intraperitoneal high dose administration of free cisplatin (150 µg) at inhibiting tumour growth.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged-Ring Compounds/chemistry , Cisplatin/pharmacology , Gelatin/chemistry , Hydrogels , Imidazoles/chemistry , Polyvinyl Alcohol/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cisplatin/chemistry , Delayed-Action Preparations , Drug Compounding , Drug Implants , Female , Humans , Injections, Intraperitoneal , Kinetics , Male , Mice , Mice, Nude , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
J Inorg Biochem ; 128: 124-30, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23954482

ABSTRACT

The sgc8c aptamer is a 41-base DNA oligonucleotide that binds to leukaemia cells with high affinity and specificity. In this work we examined the utility of this aptamer as both a delivery vehicle and an active targeting agent for an inert platinum complex [(1,10-phenathroline)(ethylenediamine)platinum(II)](2+). The aptamer forms a stem-and-loop confirmation as determined by circular dichroism. This conformation is adopted in both water and phosphate buffered saline solutions. The metal complex binds through intercalation into the aptamer's double helical stem with a binding constant of approximately 4.3 × 10(4) M(-1). Binding of the metal complex to the aptamer had a significant effect on the aptamer's global conformation, and increased its melting temperature by 28°C possibly through lengthening and stiffening of the aptamer stem. The effect of the aptamer on the metal complex's cytotoxicity and cellular uptake was determined using in vitro assays with the target leukaemia cell line CCRF-CEM and the off-target ovarian cancer cell lines A2780 and A2780cp70. The aptamer has little inherent cytotoxicity and when used to deliver the metal complex results in a significant decrease in the metal complex's cytotoxicity and uptake. The reason(s) for the poor uptake and activity may be due to the change in aptamer conformation which affects its ability to recognise leukaemia cells.


Subject(s)
Aptamers, Nucleotide/chemistry , Drug Delivery Systems/methods , Organoplatinum Compounds/administration & dosage , Phenanthrolines/chemistry , Platinum/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Aptamers, Nucleotide/genetics , Base Sequence , Cell Survival/drug effects , Circular Dichroism , Coordination Complexes/administration & dosage , Coordination Complexes/chemistry , Inhibitory Concentration 50 , Molecular Sequence Data , Molecular Structure , Nucleic Acid Conformation , Organoplatinum Compounds/chemistry
4.
Dalton Trans ; 41(37): 11330-9, 2012 Oct 07.
Article in English | MEDLINE | ID: mdl-22886151

ABSTRACT

Picoplatin is a sterically hindered mononuclear platinum drug undergoing clinical trials. The 2-methylpyridine ring provides steric hindrance to the drug, preventing attack from biological nucleophiles. BBR3464 is a trinuclear platinum drug which was recently in Phase II clinical trials, and is highly cytotoxic both in vitro and in vivo; it derives this activity through the flexible adducts it forms with DNA. In this work we sought to combine the properties of both drugs to synthesise a family of sterically hindered, dinuclear platinum complexes as potential anticancer agents. The bis-pyridyl-based ligands were synthesised through a peptide coupling reaction using diaminoalkanes of differing lengths (n = 2, 4 or 8) and 4-carboxypyridine or 2-methyl-4-carboxypyridine. The resultant dinuclear platinum complexes were synthesised by reacting two equivalents of transplatin or mono-aquated transplatin to each ligand, followed by purification by precipitation with acetone. The unprotected complexes react faster with 5'-guanosine monophosphate (drug to nucleotide ratio 1:2; t(1/2) = 2 h), glutathione (1:10, t(1/2) = 55 min) and human serum albumin (HSA) (1:1, t(1/2) = 24 h) compared to their hindered, protected equivalents (5'-guanosine monophosphate, t(1/2) = 3.5 h; glutathione = 1.7 h; HSA, t(1/2) = 110 h). The complexes were tested for in vitro cytotoxicity in the A2780 and A2780/cp70 ovarian cancer cell line. The unprotected platinum complexes were more cytotoxic than their protected derivatives, but none of the complexes were able to overcome resistance. The results provide important proof-of-concept for the development of a larger family of sterically hindered multinuclear-based platinum complexes.


Subject(s)
Antineoplastic Agents , Organoplatinum Compounds/chemistry , Ovarian Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Calorimetry, Differential Scanning , Crystallography, X-Ray , Drug Resistance, Neoplasm/drug effects , Female , Humans , Inhibitory Concentration 50 , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/pharmacokinetics , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/toxicity , Protein Binding , Tumor Cells, Cultured
5.
Metallomics ; 4(6): 561-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22610518

ABSTRACT

The cucurbit[n]uril (CB[n]) family of macrocycles has been shown to have potential in drug delivery where they are able to provide physical and chemical stability to drugs, improve drug solubility, control drug release and mask the taste of drugs. Cisplatin is a small molecule platinum-based anticancer drug that has severe dose-limiting side-effects. Cisplatin forms a host-guest complex with cucurbit[7]uril (cisplatin@CB[7]) with the platinum atom and both chlorido ligands located inside the macrocycle, with binding stabilised by four hydrogen bonds (2.15-2.44 Å). Whilst CB[7] has no effect on the in vitro cytotoxicity of cisplatin in the human ovarian carcinoma cell line A2780 and its cisplatin-resistant sub-lines A2780/cp70 and MCP1, there is a significant effect on in vivo cytotoxicity using human tumour xenografts. Cisplatin@CB[7] is just as effective on A2780 tumours compared with free cisplatin, and in the cisplatin-resistant A2780/cp70 tumours cisplatin@CB[7] markedly slows tumour growth. The ability of cisplatin@CB[7] to overcome resistance in vivo appears to be a pharmacokinetic effect. Whilst the peak plasma level and tissue distribution are the same for cisplatin@CB[7] and free cisplatin, the total concentration of circulating cisplatin@CB[7] over a period of 24 hours is significantly higher than for free cisplatin when administered at the equivalent dose. The results provide the first example of overcoming drug resistance via a purely pharmacokinetic effect rather than drug design or better tumour targeting, and demonstrate that in vitro assays are no longer as important in screening advanced systems of drug delivery.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged-Ring Compounds/pharmacology , Cisplatin/pharmacology , Drug Carriers/chemistry , Imidazoles/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Bridged-Ring Compounds/chemistry , Bridged-Ring Compounds/pharmacokinetics , Cell Line, Tumor , Cisplatin/chemistry , Cisplatin/pharmacokinetics , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Drug Carriers/therapeutic use , Drug Resistance, Neoplasm , Humans , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Mice , Mice, Nude , Models, Molecular , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Xenograft Model Antitumor Assays
6.
J Inorg Biochem ; 105(9): 1115-22, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21704583

ABSTRACT

Aquated cisplatin was added to half-generation PAMAM dendrimers and the resultant complexes were purified by centrifuge. The drug-dendrimer complexes were then characterised by 1-D and diffusion (1)H NMR and ICP-AES. The amount of drug bound was found to increase in proportion with dendrimer size: G3.5, 22 cis-{Pt(NH(3))(2)} molecules per dendrimer; G4.5, 37; G5.5, 54; and G6.5, 94, which represent only a fraction of the available binding sites on each dendrimer (68, 58, 42 and 37%, respectively). Drug release studies showed that some drug remains bound to the dendrimer even after prolonged incubation with 5'-GMP at temperatures of 60°C for over a week (percentage of drug released 18, 30, 35 and 63%, respectively). Attachment of the drug was found to decrease the radius of the dendrimers. Finally, the effect of the dendrimer on drug cytotoxicity was determined using in vitro assays with the A2780, A2780cis and A2780cp ovarian cancer cell lines. The free dendrimers display no cytotoxicity whilst the drug-dendrimer complexes showed moderate activity. In vivo activity was examined using an A2780 tumour xenograft. Cisplatin, at its maximum tolerated dose of 6 mg/kg, reduced tumour size by 33% compared to an untreated control group. The G6.5 cisplatin-dendrimer complex was administered at two doses (6 and 8 mg/kg equivalent of cisplatin). Both were well tolerated by the mice. The lower dose displayed comparable activity to cisplatin with a tumour volume reduction of 32%, but the higher dose was significantly more active than free cisplatin with a tumour reduction of 45%.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/administration & dosage , Dendrimers/chemical synthesis , Drug Carriers/chemical synthesis , Ovarian Neoplasms/drug therapy , Polyamines/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/chemistry , Cisplatin/metabolism , Dendrimers/administration & dosage , Dendrimers/metabolism , Dose-Response Relationship, Drug , Drug Carriers/administration & dosage , Drug Carriers/metabolism , Drug Dosage Calculations , Female , Guanosine Monophosphate/metabolism , Humans , Kinetics , Magnetic Resonance Spectroscopy , Mice , Mice, Nude , Ovarian Neoplasms/pathology , Particle Size , Polyamines/administration & dosage , Polyamines/metabolism , Spectrophotometry, Atomic , Xenograft Model Antitumor Assays
7.
J Am Chem Soc ; 132(13): 4678-84, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20225865

ABSTRACT

The platinum-based anticancer drugs cisplatin, carboplatin, and oxaliplatin are an important component of chemotherapy but are limited by severe dose-limiting side effects and the ability of tumors to develop resistance rapidly. These drugs can be improved through the use of drug-delivery vehicles that are able to target cancers passively or actively. In this study, we have tethered the active component of the anticancer drug oxaliplatin to a gold nanoparticle for improved drug delivery. Naked gold nanoparticles were functionalized with a thiolated poly(ethylene glycol) (PEG) monolayer capped with a carboxylate group. [Pt(1R,2R-diaminocyclohexane)(H(2)O)(2)]2NO(3) was added to the PEG surface to yield a supramolecular complex with 280 (+/-20) drug molecules per nanoparticle. The platinum-tethered nanoparticles were examined for cytotoxicity, drug uptake, and localization in the A549 lung epithelial cancer cell line and the colon cancer cell lines HCT116, HCT15, HT29, and RKO. The platinum-tethered nanoparticles demonstrated as good as, or significantly better, cytotoxicity than oxaliplatin alone in all of the cell lines and an unusual ability to penetrate the nucleus in the lung cancer cells.


Subject(s)
Antineoplastic Agents/metabolism , Drug Carriers/chemistry , Drug Delivery Systems , Gold/chemistry , Metal Nanoparticles/chemistry , Organoplatinum Compounds/metabolism , Organoplatinum Compounds/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Cell Death/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Drug Carriers/chemical synthesis , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/toxicity , Oxaliplatin , Polyethylene Glycols/chemistry , Structure-Activity Relationship
8.
Clin Cancer Res ; 14(3): 804-10, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18245542

ABSTRACT

PURPOSE: To determine the safety, dose-limiting toxicity, maximum tolerated dose, and pharmacokinetic and pharmacodynamic profiles of the novel hydroxamate histone deacetylase inhibitor belinostat (previously named PXD101) in patients with advanced refractory solid tumors. EXPERIMENTAL DESIGN: Sequential dose-escalating cohorts of three to six patients received belinostat administered as a 30-min i.v. infusion on days 1 to 5 of a 21-day cycle. Pharmacokinetic variables were evaluated at all dose levels. Pharmacodynamic measurements included acetylation of histones extracted from peripheral blood mononuclear cells, caspase-dependent cleavage of cytokeratin-18, and interleukin-6 levels. RESULTS: Forty-six patients received belinostat at one of six dose levels (150-1,200 mg/m(2)/d). Dose-limiting toxicities were grade 3 fatigue (one patient at 600 mg/m(2); one patient at 1,200 mg/m(2)), grade 3 diarrhea combined with fatigue (one patient at 1,200 mg/m(2)), grade 3 atrial fibrillation (one patient at 1,200 mg/m(2); one patient at 1,000 mg/m(2)), and grade 2 nausea/vomiting leading to inability to complete a full 5-day cycle (two patients at 1,000 mg/m(2)). The maximum tolerated dose was 1,000 mg/m(2)/d. I.v. belinostat displayed linear pharmacokinetics with respect to C(max) and AUC. The intermediate elimination half-life was 0.3 to 1.3 h and was independent of dose. Histone H4 hyperacetylation was observed after each infusion and was sustained for 4 to 24 h in a dose-dependent manner. Increases in interleukin-6 levels were detected following belinostat treatment. Stable disease was observed in a total of 18 (39%) patients, including 15 treated for > or =4 cycles, and this was associated with caspase-dependent cleavage of cytokeratin-18. Of the 24 patients treated at the maximum tolerated dose (1,000 mg/m(2)/d), 50% achieved stable disease. CONCLUSIONS: I.v. belinostat is well tolerated, exhibits dose-dependent pharmacodynamic effects, and has promising antitumor activity.


Subject(s)
Antineoplastic Agents/toxicity , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/toxicity , Histone Deacetylase Inhibitors , Histone Deacetylases/pharmacokinetics , Hydroxamic Acids/toxicity , Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents/administration & dosage , Drug Administration Schedule , Enzyme Inhibitors/administration & dosage , Female , Humans , Infusions, Intravenous , Life Expectancy , Male , Middle Aged , Neoplasms/pathology , Patient Selection , Sulfonamides
9.
Proc Natl Acad Sci U S A ; 104(45): 17777-82, 2007 Nov 06.
Article in English | MEDLINE | ID: mdl-17968018

ABSTRACT

Understanding the pathways that are targeted by cancer drugs is instrumental for their rational use in a clinical setting. Inhibitors of histone deacetylases (HDACI) selectively inhibit proliferation of malignant cells and are used for the treatment of cancer, but their cancer selectivity is understood poorly. We conducted a functional genetic screen to address the mechanism(s) of action of HDACI. We report here that ectopic expression of two genes that act on retinoic acid (RA) signaling can cause resistance to growth arrest and apoptosis induced by HDACI of different chemical classes: the retinoic acid receptor alpha (RARalpha) and preferentially expressed antigen of melanoma (PRAME), a repressor of RA signaling. Treatment of cells with HDACI induced RA signaling, which was inhibited by RARalpha or PRAME expression. Conversely, RAR-deficient cells and PRAME-knockdown cells show enhanced sensitivity to HDACI in vitro and in mouse xenograft models. Finally, a combination of RA and HDACI acted synergistically to activate RA signaling and inhibit tumor growth. These experiments identify the RA pathway as a rate-limiting target of HDACI and suggest strategies to enhance the therapeutic efficacy of HDACI.


Subject(s)
Genetic Testing/methods , Histone Deacetylase Inhibitors , Histone Deacetylases/genetics , Tretinoin/physiology , Animals , Cell Division/drug effects , Colony-Forming Units Assay , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Fibroblasts/physiology , Humans , Mice , Neoplasm Transplantation , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/physiology , Retinoic Acid Receptor alpha , Transplantation, Heterologous , Tretinoin/pharmacology
10.
J Clin Oncol ; 25(29): 4603-9, 2007 Oct 10.
Article in English | MEDLINE | ID: mdl-17925555

ABSTRACT

PURPOSE: The DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (decitabine) induces DNA demethylation and re-expression of epigenetically silenced genes, and increases carboplatin sensitivity of tumor xenograft models. We designed a clinical study to determine the feasibility of delivering a dose of decitabine, combined with carboplatin, that would be capable of producing equivalent biologic effects in patients with solid tumors. PATIENTS AND METHODS: In a two-stage design, 33 patients received escalating doses of decitabine administered as a 6-hour infusion on day 1 followed by carboplatin, area under the concentration-time curve (AUC) 5 (cohort 1) and AUC 6 (cohort 2), on day 8 of a 28-day cycle. Pharmacodynamic analyses included 5-methyl-2'-deoxycytidine levels, MAGE1A CpG island methylation, and fetal hemoglobin (HbF) expression. RESULTS: The major toxicity was myelosuppression. Dose limiting toxicities, prolonged grade 4 neutropenia (one patient), and sepsis and grade 3 anorexia/fatigue (one patient), were seen in two of four patients treated with decitabine 135 mg/m2 and carboplatin AUC 5. Dose limiting toxicity comprising neutropenic sepsis (one patient) and grade 3 fatigue (one patient) was seen in two of 10 patients treated at decitabine 90 mg/m2 and carboplatin AUC 6. Decitabine induced dose-dependent, reversible demethylation in peripheral-blood cells (PBCs) maximally at day 10. Furthermore, decitabine 90 mg/m2 induced demethylation of the MAGE1A CpG island in PBCs, buccal cells, and tumor biopsies, as well as elevation of HbF expression. CONCLUSION: Decitabine can be combined safely with carboplatin at a dose and schedule that causes epigenetic changes equivalent to or greater than that observed in mice with carboplatin-sensitized xenografts. The recommended dose/schedule for phase II trials is decitabine 90 mg/m2 (day 1) followed by carboplatin AUC 6 (day 8) every 28 days.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/analogs & derivatives , Carboplatin/pharmacology , DNA Modification Methylases/antagonists & inhibitors , Neoplasms/drug therapy , Adult , Aged , Area Under Curve , Azacitidine/administration & dosage , Azacitidine/pharmacology , Carboplatin/administration & dosage , Cohort Studies , CpG Islands , DNA Methylation , Decitabine , Female , Humans , Male , Middle Aged
11.
J Med Chem ; 48(21): 6690-5, 2005 Oct 20.
Article in English | MEDLINE | ID: mdl-16220985

ABSTRACT

A novel series of 1,4-disubstituted aminoanthraquinones were prepared by ipso-displacement of 1,4-difluoro-5,8-dihydroxyanthraquinones by hydroxylated piperidinyl- or pyrrolidinylalkylamino side chains. One aminoanthraquinone (13) was further derivatized to a chloropropylamino analogue by treatment with triphenylphosphine-carbon tetrachloride. The compounds were evaluated in the A2780 ovarian cancer cell line and its cisplatin-resistant variants (A2780/cp70 and A2780/MCP1). The novel anthraquinones were shown to possess up to 5-fold increased potency against the cisplatin-resistant cells compared to the wild-type cells. Growth curve analysis of the hydroxyethylaminoanthraquinone 8 in the osteosarcoma cell line U-2 OS showed that the cell cycle is not frozen, rather there is a late cell cycle arrest consistent with the action of a DNA-damaging topoisomerase II inhibitor. Accumulative apoptotic events, using time lapse photography, indicate that 8 is capable of fully engaging cell cycle arrest pathways in G2 in the absence of early apoptotic commitment. 8 and its chloropropyl analogue 13 retained significant activity against human A2780/cp70 xenografted tumors in mice.


Subject(s)
Anthraquinones/chemical synthesis , Antineoplastic Agents/chemical synthesis , Cisplatin/pharmacology , Animals , Anthraquinones/chemistry , Anthraquinones/pharmacology , Antigens, Neoplasm , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , DNA Topoisomerases, Type II , DNA-Binding Proteins/antagonists & inhibitors , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, Nude , Ovarian Neoplasms , Structure-Activity Relationship , Topoisomerase II Inhibitors , Transplantation, Heterologous
12.
Carcinogenesis ; 26(11): 1856-67, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15958521

ABSTRACT

The incidence of melanoma is increasing rapidly, with advanced lesions generally failing to respond to conventional chemotherapy. Here, we utilized DNA microarray-based gene expression profiling techniques to identify molecular determinants of melanoma progression within a unique panel of isogenic human melanoma cell lines. When a poorly tumorigenic cell line, derived from an early melanoma, was compared with two increasingly aggressive derivative cell lines, the expression of 66 genes was significantly changed. A similar pattern of differential gene expression was found with an independently derived metastatic cell line. We further examined these melanoma progression-associated genes via use of a tailored TaqMan Low Density Array (LDA), representing the majority of genes within our cohort of interest. Considerable concordance was seen between the transcriptomic profiles determined by DNA microarray and TaqMan LDA approaches. A range of novel markers were identified that correlated here with melanoma progression. Most notable was TSPY, a Y chromosome-specific gene that displayed extensive down-regulation in expression between the parental and derivative cell lines. Examination of a putative CpG island within the TSPY gene demonstrated that this region was hypermethylated in the derivative cell lines, as well as metastatic melanomas from male patients. Moreover, treatment of the derivative cell lines with the DNA methyltransferase inhibitor, 2'-deoxy-5-azacytidine (DAC), restored expression of the TSPY gene to levels comparable with that found in the parental cells. Additional DNA microarray studies uncovered a subset of 13 genes from the above-mentioned 66 gene cohort that displayed re-activation of expression following DAC treatment, including TSPY, CYBA and MT2A. DAC suppressed tumor cell growth in vitro. Moreover, systemic treatment of mice with DAC attenuated growth of melanoma xenografts, with consequent re-expression of TSPY mRNA. Overall, our data support the hypothesis that multiple genes are targeted, either directly or indirectly, by DNA hypermethylation during melanoma progression.


Subject(s)
DNA Methylation , Gene Expression Regulation, Neoplastic , Melanoma, Experimental/genetics , Skin Neoplasms/genetics , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Azacitidine/therapeutic use , Biomarkers, Tumor , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Decitabine , Disease Progression , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Epigenesis, Genetic , Female , Gene Expression Profiling , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/prevention & control , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Skin Neoplasms/prevention & control , Skin Neoplasms/secondary , Transplantation, Heterologous , Tumor Cells, Cultured
13.
Expert Rev Anticancer Ther ; 4(4): 501-10, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15270655

ABSTRACT

Genes involved in all aspects of tumor development and growth can become aberrantly methylated in tumor cells, including genes involved in apoptosis and cell cycle regulation. Decitabine, 2'-deoxy-5-azacytidine, can inhibit DNA methyltransferases and reverse epigenetic silencing of aberrantly methylated genes. Nucleoside DNA methyltransferase inhibitors, such as decitabine, have been reported to have antitumor activity, especially against hematologic malignancies. Such demethylating agents have been proposed to reactivate tumor suppressor genes aberrantly methylated in tumor cells, leading to inhibition of tumor growth. An important consequence of this is that, unlike conventional cytotoxic agents, it may be best to use such drugs at concentrations lower than the maximum tolerated dose and in a manner dependent on their demethylating activity. Furthermore, synergistic activity with other types of investigational epigenetic therapies and existing chemotherapies opens the possibility of rational combinations and scheduling of these agents based on their biologic activity.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/pharmacokinetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Azacitidine/pharmacokinetics , DNA Methylation , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Hematologic Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Cycle/drug effects , Cell Differentiation/drug effects , Decitabine , Humans
16.
Mol Cancer Ther ; 2(8): 721-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12939461

ABSTRACT

Histone acetylation has a central role in the control of gene expression, influencing transcriptional control of many genes, including tumor suppressor genes. PXD101 is a novel hydroxamate-type inhibitor of histone deacetylase activity that inhibits histone deacetylase activity in HeLa cell extracts with an IC(50) of 27 nM and induces a concentration-dependent (0.2-5 micro M) increase in acetylation of histone H4 in tumor cell lines. PXD101 is cytotoxic in vitro in a number of tumor cell lines with IC(50)s in the range 0.2-3.4 micro M as determined by a clonogenic assay and induces apoptosis. Treatment of nude mice bearing human ovarian and colon tumor xenografts with PXD101 (10-40 mg/kg/day i.p.) daily for 7 days causes a significant dose-dependent growth delay with no obvious signs of toxicity to the mice. Growth delay is also observed for xenografts of cisplatin-resistant ovarian tumor cells. A marked increase in acetylation of H4 is detected in blood and tumor of mice 3 h after treatment with PXD101. The inhibition of growth of human tumor xenografts in mice, with no apparent toxicity, suggests that PXD101 has potential as a novel antitumor agent. Furthermore, the ability to measure histone acetylation in blood samples could provide a suitable pharmacodynamic end point to monitor drug activity.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Acetylation/drug effects , Animals , Apoptosis , Cell Line, Tumor , Dose-Response Relationship, Drug , HeLa Cells , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Hydroxamic Acids , Mice , Mice, Nude , Neoplasms, Experimental/drug therapy , Structure-Activity Relationship , Sulfonamides , Transplantation, Heterologous , Xenograft Model Antitumor Assays
17.
Mol Cancer Ther ; 2(7): 607-10, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12883032

ABSTRACT

Chloroethylaminoanthraquinones are described with intercalating and alkylating capacity that potentially covalently cross-link topoisomerase II (topo II) to DNA. These compounds have potent cytotoxic activity (IC(50) = 0.9-7.6 nM) against the A2780 human ovarian carcinoma cell line. Hydroxyethylaminoanthraquinones also reported in this paper have similar IC(50) values (0.7-1.7 nM) in the same cell line. Alchemix (ZP281M, 1-(2-[N,N-bis(2-chloroethyl)amino]ethylamino)-4-(2-[N,N-(dimethyl)amino]ethylamino)-5,8-dihydroxy-9,10-anthracenedione), an alkylating anthraquinone, retains excellent antitumor activity in Adriamycin-resistant (2780AD) and cisplatin-resistant (2780/cp70) cell lines in vitro and in vivo. This indicates that Alchemix can evade both P-glycoprotein efflux pump and DNA mismatch repair-mediated resistance. In treated cells, Alchemix was shown to preferentially induce drug-stabilized covalent bound topo IIalpha-DNA complexes over topo IIbeta-DNA complexes.


Subject(s)
Anthracyclines/pharmacology , Anthraquinones/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Ovarian Neoplasms/drug therapy , Animals , Antigens, Neoplasm , DNA/metabolism , DNA Repair , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins , Female , Humans , In Vitro Techniques , Mice , Mice, Nude , Ovarian Neoplasms/metabolism , Tumor Cells, Cultured
18.
Oncogene ; 22(3): 370-80, 2003 Jan 23.
Article in English | MEDLINE | ID: mdl-12545158

ABSTRACT

Reactivation of telomerase maintains telomere function and is considered critical to immortalization in most human cancer cells. Elevation of telomerase expression in cancer cells is highly specific: transcription of both RNA (hTR) and protein (hTERT) components is strongly upregulated in cancer cells relative to normal cells. Therefore, telomerase promoters may be useful in cancer gene therapy by selectively expressing suicide genes in cancer cells and not normal cells. One example of suicide gene therapy is the bacterial nitroreductase (NTR) gene, which bioactivates the prodrug CB1954 into an active cytotoxic alkylating agent. We describe construction of adenovirus vectors harbouring the bacterial NTR gene under control of the hTR or hTERT promoters. Western blot analysis of NTR expression in normal and cancer cells infected with adenoviral vectors showed cancer cell-specific nitroreductase expression. Infection with adenoviral telomerase-NTR constructs in a panel of seven cancer cell lines resulted in up to 18-fold sensitization to the prodrug CB1954, an effect that was retained in two drug-resistant ovarian lines. Importantly, no sensitization was observed with either promoter in any of the four normal cell strains. Finally, an efficacious effect was observed in cervical and ovarian xenograft models following single intratumoural injection with low doses of vector, followed by injection with CB1954.


Subject(s)
Adenoviridae/genetics , Bacterial Proteins/genetics , Genetic Therapy/methods , Genetic Vectors/pharmacology , Neoplasms/therapy , Nitroreductases/genetics , Telomerase/drug effects , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Animals , Antineoplastic Agents/pharmacology , Aziridines/pharmacology , Bacterial Proteins/metabolism , Base Sequence , DNA-Binding Proteins , Drug Resistance, Neoplasm , Female , Genetic Vectors/genetics , Humans , Male , Mice , Mice, Nude , Molecular Sequence Data , Neoplasms/genetics , Nitroreductases/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Promoter Regions, Genetic , Telomerase/genetics , Telomerase/metabolism , Tumor Cells, Cultured , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...