Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters










Publication year range
1.
RSC Chem Biol ; 4(10): 736-741, 2023 Oct 04.
Article in English | MEDLINE | ID: mdl-37799580

ABSTRACT

It is 40 years since the publication of the patent that announced the imidazotetrazines temozolomide and mitozolomide to the world and 30 since the discovery that they function as prodrugs of alkyldiazonium reactive intermediates. Temozolomide combined with radiation is established as the first-line treatment for glioma but despite the attentions of the inventors and others, further examples of this intriguing ring system have yet to enter the clinic.

2.
Eur J Med Chem ; 257: 115507, 2023 Sep 05.
Article in English | MEDLINE | ID: mdl-37262998

ABSTRACT

We describe the design, organic synthesis, and characterization, including X-ray crystallography, of a series of novel analogues of the clinically used antitumor agent temozolomide, together with their in vitro biological evaluation. The work has resulted in the discovery of a new series of anticancer imidazotetrazines that offer the potential to overcome the resistance mounted by tumors against temozolomide. The rationally designed compounds that incorporate a propargyl alkylating moiety and a thiazole ring as isosteric replacement for a carboxamide, are readily synthesized (gram-scale), exhibit defined solid-state structures, and enhanced growth-inhibitory activity against human tumor cell lines, including MGMT-expressing and MMR-deficient lines, molecular features that confer tumor resistance. The cell proliferation data were confirmed by clonogenic cell survival assays, and DNA flow cytometry analysis was undertaken to determine the effects of new analogues on cell cycle progression. Detailed 1H NMR spectroscopic studies showed that the new agents are stable in solution, and confirmed their mechanism of action. The propargyl and thiazole substituents significantly improve potency and physicochemical, drug metabolism and permeability properties, suggesting that the thiazole 13 should be prioritized for further preclinical evaluation.


Subject(s)
Antineoplastic Agents , Brain Neoplasms , Glioblastoma , Humans , Temozolomide/pharmacology , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Brain Neoplasms/drug therapy , Cell Line, Tumor , Thiazoles/pharmacology , Thiazoles/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Drug Resistance, Neoplasm , Glioblastoma/drug therapy
3.
ACS Appl Mater Interfaces ; 13(30): 35266-35280, 2021 Aug 04.
Article in English | MEDLINE | ID: mdl-34310112

ABSTRACT

The lack of clinical response to the alkylating agent temozolomide (TMZ) in pediatric diffuse midline/intrinsic pontine glioma (DIPG) has been associated with O6-methylguanine-DNA-methyltransferase (MGMT) expression and mismatch repair deficiency. Hence, a potent N(3)-propargyl analogue (N3P) was derived, which not only evades MGMT but also remains effective in mismatch repair deficient cells. Due to the poor pharmacokinetic profile of N3P (t1/2 < 1 h) and to bypass the blood-brain barrier, we proposed convection enhanced delivery (CED) as a method of administration to decrease dose and systemic toxicity. Moreover, to enhance N3P solubility, stability, and sustained distribution in vivo, either it was incorporated into an apoferritin (AFt) nanocage or its sulfobutyl ether ß-cyclodextrin complex was loaded into nanoliposomes (Lip). The resultant AFt-N3P and Lip-N3P nanoparticles (NPs) had hydrodynamic diameters of 14 vs 93 nm, icosahedral vs spherical morphology, negative surface charge (-17 vs -34 mV), and encapsulating ∼630 vs ∼21000 N3P molecules per NP, respectively. Both NPs showed a sustained release profile and instant uptake within 1 h incubation in vitro. In comparison to the naked drug, N3P NPs demonstrated stronger anticancer efficacy against 2D TMZ-resistant DIPG cell cultures [IC50 = 14.6 (Lip-N3P) vs 32.8 µM (N3P); DIPG-IV) and (IC50 = 101.8 (AFt-N3P) vs 111.9 µM (N3P); DIPG-VI)]. Likewise, both N3P-NPs significantly (P < 0.01) inhibited 3D spheroid growth compared to the native N3P in MGMT+ DIPG-VI (100 µM) and mismatch repair deficient DIPG-XIX (50 µM) cultures. Interestingly, the potency of TMZ was remarkably enhanced when encapsulated in AFt NPs against DIPG-IV, -VI, and -XIX spheroid cultures. Dynamic PET scans of CED-administered zirconium-89 (89Zr)-labeled AFt-NPs in rats also demonstrated substantial enhancement over free 89Zr radionuclide in terms of localized distribution kinetics and retention within the brain parenchyma. Overall, both NP formulations of N3P represent promising approaches for treatment of TMZ-resistant DIPG and merit the next phase of preclinical evaluation.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Drug Carriers/chemistry , Glioma/drug therapy , Nanoparticles/chemistry , Temozolomide/analogs & derivatives , Temozolomide/therapeutic use , Animals , Apoferritins/chemistry , Cell Line, Tumor , Humans , Liposomes/chemistry , Male , Rats, Wistar , Spheroids, Cellular/drug effects , beta-Cyclodextrins/chemistry
4.
ACS Appl Mater Interfaces ; 12(11): 12609-12617, 2020 Mar 18.
Article in English | MEDLINE | ID: mdl-32073826

ABSTRACT

Glioblastoma multiforme (GBM) is a grade IV astrocytoma, which is the most aggressive form of brain tumor. The standard of care for this disease includes surgery, radiotherapy and temozolomide (TMZ) chemotherapy. Poor accumulation of TMZ at the tumor site, tumor resistance to drug, and dose-limiting bone marrow toxicity eventually reduce the success of this treatment. Herein, we have encapsulated >500 drug molecules of TMZ into the biocompatible protein nanocage, apoferritin (AFt), using a "nanoreactor" method (AFt-TMZ). AFt is internalized by transferrin receptor 1-mediated endocytosis and is therefore able to facilitate cancer cell uptake and enhance drug efficacy. Following encapsulation, the protein cage retained its morphological integrity and surface charge; hence, its cellular recognition and uptake are not affected by the presence of this cargo. Additional benefits of AFt include maintenance of TMZ stability at pH 5.5 and drug release under acidic pH conditions, encountered in lysosomal compartments. MTT assays revealed that the encapsulated agents displayed significantly increased antitumor activity in U373V (vector control) and, remarkably, the isogenic U373M (MGMT expressing TMZ-resistant) GBM cell lines, with GI50 values <1.5 µM for AFt-TMZ, compared to 35 and 376 µM for unencapsulated TMZ against U373V and U373M, respectively. The enhanced potency of AFt-TMZ was further substantiated by clonogenic assays. Potentiated G2/M cell cycle arrest following exposure of cells to AFt-TMZ indicated an enhanced DNA damage burden. Indeed, increased O6-methylguanine (O6-MeG) adducts in cells exposed to AFt-TMZ and subsequent generation of γH2AX foci support the hypothesis that AFt significantly enhances the delivery of TMZ to cancer cells in vitro, overwhelming the direct O6-MeG repair conferred by MGMT. We have additionally encapsulated >500 molecules of the N3-propargyl imidazotetrazine analog (N3P), developed to combat TMZ resistance, and demonstrated significantly enhanced activity of AFt-N3P against GBM and colorectal carcinoma cell lines. These studies support the use of AFt as a promising nanodelivery system for targeted delivery, lysosomal drug release, and enhanced imidazotetrazine potency for treatment of GBM and wider-spectrum malignancies.


Subject(s)
Antineoplastic Agents, Alkylating , Apoferritins/chemistry , Brain Neoplasms/metabolism , Nanostructures/chemistry , Temozolomide , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/pharmacokinetics , Antineoplastic Agents, Alkylating/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Delivery Systems , Glioblastoma/metabolism , Humans , Temozolomide/analogs & derivatives , Temozolomide/chemistry , Temozolomide/pharmacokinetics , Temozolomide/pharmacology
5.
J Cell Mol Med ; 24(2): 1750-1759, 2020 01.
Article in English | MEDLINE | ID: mdl-31876059

ABSTRACT

Stomach cancer is the fourth most common cancer worldwide. Identification of novel molecular therapeutic targets and development of novel treatments are critical. Against a panel of gastric carcinoma cell lines, the activity of 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (5F 203) was investigated. Adopting RT-PCR, Western blot and immunohistochemical techniques, we sought to determine molecular pharmacodynamic (PD) markers of sensitivity and investigate arylhydrocarbon (AhR) receptor-mediated signal transduction activation by 5F 203. Potent (IC50  ≤ 0.09 µmol/L), selective (>250-fold) in vitro antitumour activity was observed in MKN-45 and AGS carcinoma cells. Exposure of MKN-45 cells to 5F 203 triggered cytosolic AhR translocation to nuclei, inducing CYP1A1 (>50-fold) and CYP2W1 (~20-fold) transcription and protein (CYP1A1 and CYP2W1) expression. G2/M arrest and γH2AX expression preceded apoptosis, evidenced by PARP cleavage. In vivo, significant (P < .01) 5F 203 efficacy was observed against MKN-45 and AGS xenografts. In mice-bearing 5F 203-sensitive MKN-45 and 5F 203-insensitive BGC-823 tumours in opposite flanks, CYP1A1, CYP2W1 and γH2AX protein in MKN-45 tumours only following treatment of mice with 5F 203 (5 mg/kg) revealed PD biomarkers of sensitivity. 5F 203 evokes potent, selective antitumour activity in vitro and in vivo in human gastric cancer models. It triggers AhR signal transduction, CYP-catalysed bioactivation to electrophilic species causing lethal DNA double-strand breaks exclusively in sensitive cells. 5F 203 represents a novel therapeutic agent with a mechanism of action distinct from current clinical drugs, exploiting novel molecular targets pertinent to gastric tumourigenesis: AhR, CYP1A1 and CYP2W1. PD markers of 5F 203 sensitivity that could guide patient selection have been identified.


Subject(s)
Antineoplastic Agents/therapeutic use , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Thiazoles/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , DNA Damage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Space/metabolism , Mice, Inbred BALB C , Mice, Nude , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Thiazoles/chemistry , Thiazoles/pharmacology
6.
Front Oncol ; 9: 485, 2019.
Article in English | MEDLINE | ID: mdl-31263673

ABSTRACT

Temozolomide (TMZ) is the standard of care chemotherapeutic agent used in the treatment of glioblastoma multiforme. Cytotoxic O6-methylguaine lesions formed by TMZ are repaired by O6-methyl-guanine DNA methyltransferase (MGMT), a DNA repair protein that removes alkyl groups located at the O6-position of guanine. Response to TMZ requires low MGMT expression and functional mismatch repair. Resistance to TMZ conferred by MGMT, and tolerance to O6-methylguanine lesions conferred by deficient MMR severely limit TMZ clinical applications. Therefore, development of new TMZ derivatives that can overcome TMZ-resistance is urgent. In this study, we investigated the anti-tumor mechanism of action of two novel TMZ analogs: C8-imidazolyl (377) and C8-methylimidazole (465) tetrazines. We found that analogs 377 and 465 display good anticancer activity against MGMT-overexpressing glioma T98G and MMR deficient colorectal carcinoma HCT116 cell lines with IC50 value of 62.50, 44.23, 33.09, and 25.37 µM, respectively. Analogs induce cell cycle arrest at G2/M, DNA double strand break damage and apoptosis irrespective of MGMT and MMR status. It was established that analog 377, similar to TMZ, is able to ring-open and hydrolyze under physiological conditions, and its intermediate product is more stable than MTIC. Moreover, DNA adducts of 377 with calf thymus DNA were identified: N7-methylguanine, O6-methylguanine, N3-methyladenine, N3-methylthymine, and N3-methylcytidine deoxynucleotides. We conclude that C8 analogs of TMZ share a mechanism of action similar to TMZ and are able to methylate DNA generating O6-methylguanine adducts, but unlike TMZ are able at least in part to thwart MGMT- and MMR-mediated resistance.

7.
Cancer Drug Resist ; 2(4): 1018-1031, 2019.
Article in English | MEDLINE | ID: mdl-35582280

ABSTRACT

Glioblastoma multiforme is the most common and lethal brain tumour-type. The current standard of care includes Temozolomide (TMZ) chemotherapy. However, inherent and acquired resistance to TMZ thwart successful treatment. The direct repair protein methylguanine DNA methyltransferase (MGMT) removes the cytotoxic O6-methylguanine (O6-MeG) lesion delivered by TMZ and so its expression by tumours confers TMZ-resistance. DNA mismatch repair (MMR) is essential to process O6-MeG adducts and MMR-deficiency leads to tolerance of lesions, resistance to TMZ and further DNA mutations. In this article, two strategies to overcome TMZ resistance are discussed: (1) synthesis of imidazotetrazine analogues - designed to retain activity in the presence of MGMT or loss of MMR; (2) preparation of imidazotetrazine-nanoparticles to deliver TMZ preferably to the brain and tumour site. Our promising results encourage belief in a future where better prognoses exist for patients diagnosed with this devastating disease.

8.
Medchemcomm ; 9(3): 545-553, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-30108945

ABSTRACT

A series of 3-(benzyl-substituted)-imidazo[5,1-d]-1,2,3,5-tetrazines (13) and related derivatives with 3-heteromethyl groups has been synthesised and screened for growth-inhibitory activity in vitro against two pairs of glioma cell lines with temozolomide-sensitive and -resistant phenotypes dependent on the absence/presence of the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT). In general the compounds had low inhibitory activity with GI50 values >50 µM against both sets of cell lines. Two silicon-containing derivatives, the TMS-methylimidazotetrazine (9) and the SEM-analogue (10), showed interesting differences: compound (9) had a profile very similar to that of temozolomide with the MGMT+ cell lines being 5 to 10-fold more resistant than MGMT- isogenic partners; the SEM-substituted compound (10) showed potency across all cell lines irrespective of their MGMT status.

9.
J Cell Biochem ; 119(7): 5350-5358, 2018 07.
Article in English | MEDLINE | ID: mdl-29331023

ABSTRACT

The efficacy of temozolomide (TMZ) treatment for cancers is currently limited by inherent or the development of resistance, particularly, but not exclusively, due to the expression of the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT) in a significant proportion of tumors. We have found that TMZ analog C8-methyl imidazole tetrazine (PMX 465) displayed good anticancer activity against the colorectal carcinoma HCT116 cells which are MGMT-overexpressing and mismatch repair (MMR)-deficient. In this study, we found that PMX 465 could downregulate the expression of MGMT in HCT116 cells at the protein and mRNA levels. We found that PMX 465 could reduce MGMT expression by increasing the binding of wild-type p53 to the MGMT promoter and reducing the binding of Sp1 to the MGMT promoter.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Colorectal Neoplasms/metabolism , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Temozolomide/analogs & derivatives , Temozolomide/pharmacology , Tumor Suppressor Proteins/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Down-Regulation , HCT116 Cells , Humans , Promoter Regions, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
10.
Org Biomol Chem ; 14(16): 3889-905, 2016 Apr 28.
Article in English | MEDLINE | ID: mdl-27035072

ABSTRACT

The imidazole ring is widespread in biologically active compounds, and hence imidazole-containing scaffolds are useful starting points for drug discovery programmes. We report the synthesis of a series of novel imidazole-containing compounds fused with either phenanthrene or phenanthroline, which show enhanced growth inhibitory potency against human colon, breast and melanoma cancer cell lines, as well as evidence of inhibition of the molecular chaperone heat shock protein 70 (Hsp70) pathway in cells, as shown by depletion of downstream oncogenic client proteins of the Hsp90 chaperone pathway, and induction of apoptosis.


Subject(s)
Apoptosis/drug effects , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Imidazoles/chemistry , Imidazoles/pharmacology , Phenanthrolines/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Humans , Phenanthrolines/pharmacology
11.
Bioorg Med Chem ; 23(21): 6891-9, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26474663

ABSTRACT

Potent, selective antitumour AhR ligands 5F 203 and GW 610 are bioactivated by CYPs 1A1 and 2W1. Herein we reason that DNA adducts' generation resulting in lethal DNA double strand breaks (DSBs) underlies benzothiazoles' activity. Treatment of sensitive carcinoma cell lines with GW 610 generated co-eluting DNA adducts (R(2)>0.7). Time-dependent appearance of γ-H2AX foci revealed subsequent DNA double strand breaks. Propensity for systemic toxicity of benzothiazoles steered development of prodrugs' hydrogels for localised delivery. Clinical applications of targeted therapies include prevention or treatment of recurrent disease after surgical resection of solid tumours. In vitro evaluation of 5F 203 prodrugs' activity demonstrated nanomolar potency against MCF-7 breast and IGROV-1 ovarian carcinoma cell lines.


Subject(s)
Antineoplastic Agents/chemical synthesis , DNA Adducts/analysis , Hydrogels/chemistry , Prodrugs/chemical synthesis , Thiazoles/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzothiazoles/chemical synthesis , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , DNA Adducts/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation/drug effects , Histones/metabolism , Humans , Microscopy, Confocal , Prodrugs/chemistry , Prodrugs/pharmacology , Resveratrol , Stilbenes/chemistry , Thiazoles/chemical synthesis , Thiazoles/pharmacology
12.
Oncology ; 88(1): 28-48, 2015.
Article in English | MEDLINE | ID: mdl-25277441

ABSTRACT

Glioblastoma multiforme (GBM) treatment includes temozolomide (TMZ) chemotherapy. O6-Methylguanine lesions are repaired by methylguanine-DNA methyltransferase (MGMT). Response to TMZ requires low MGMT and functional mismatch repair (MMR); resistance, conferred by MGMT or MMR deficiency, represents a barrier to successful treatment. TMZ analogs were synthesized, substituting N3-methyl with propargyl (1) or sulfoxide (2). MTT assays were conducted in SNB19 and U373 isogenic glioma cell lines (V = vector control; M = MGMT-transfected). TMZ potency was reduced >5-fold in SNB19M and U373M cells; in contrast, MGMT-expressing cells were equisensitive as vector controls to analogs 1 and 2 . GI50 values <50 µM of analogs 1 or 2 were detected in V cells possessing acquired TMZ resistance: SNB19VR (hMSH6 loss) and U373VR (MGMT upregulation). Analogs 1 and 2 inhibited MMR-deficient colorectal carcinoma cell growth (irrespective of p53); G2/M cell cycle arrest preceded apoptosis. γH2AX foci inferred the generation of DNA double-strand breaks by analogs 1 and 2 . Acridine orange-stained vesicles, intracellular punctate GFP-LC3 protein and double-membraned autophagosomes indicate that TMZ, 1 and 2 induce autophagy in apoptotis-resistant GBM cells. Analogs 1 and 2 elicit in vitro antitumor activity irrespective of MGMT, MMR and p53. Such imidazotetrazines may treat MGMT+ GBM and possess broader spectrum activity causing apoptosis and autophagy in malignancies which evade apoptosis.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , DNA Mismatch Repair/drug effects , DNA Modification Methylases/analysis , DNA Repair Enzymes/analysis , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Tumor Suppressor Proteins/analysis , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Breaks, Double-Stranded , Dacarbazine/pharmacology , Glioblastoma/pathology , Histones/analysis , Humans , Temozolomide , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/physiology
13.
Chem Commun (Camb) ; 50(96): 15202-5, 2014 Dec 14.
Article in English | MEDLINE | ID: mdl-25338751

ABSTRACT

Biophysical studies of ligand interactions with three human telomeric repeat sequences (d(AGGG(TTAGGG)n, n = 3, 7 and 11)) show that an oxazole-based 'click' ligand, which induces parallel folded quadruplexes, preferentially stabilises longer telomeric repeats providing evidence for selectivity in binding at the interface between tandem quadruplex motifs.


Subject(s)
G-Quadruplexes , Ligands , Telomere/chemistry , Base Sequence , Binding Sites , Circular Dichroism , Click Chemistry , Humans , Molecular Dynamics Simulation , Nucleic Acid Conformation , Oxazoles/chemistry
14.
PLoS One ; 9(1): e86187, 2014.
Article in English | MEDLINE | ID: mdl-24454961

ABSTRACT

BACKGROUND: Telomeric 3' overhangs can fold into a four-stranded DNA structure termed G-quadruplex (G4), a formation which inhibits telomerase. As telomerase activation is crucial for telomere maintenance in most cancer cells, several classes of G4 ligands have been designed to directly disrupt telomeric structure. METHODS: We exposed brain tumor cells to the G4 ligand 3,11-difluoro-6,8,13-trimethyl-8H-quino[4,3,2-kl]acridinium methosulfate (RHPS4) and investigated proliferation, cell cycle dynamics, telomere length, telomerase activity and activated c-Myc levels. RESULTS: Although all cell lines tested were sensitive to RHPS4, PFSK-1 central nervous system primitive neuroectodermal cells, DAOY medulloblastoma cells and U87 glioblastoma cells exhibited up to 30-fold increased sensitivity compared to KNS42 glioblastoma, C6 glioma and Res196 ependymoma cells. An increased proportion of S-phase cells were observed in medulloblastoma and high grade glioma cells whilst CNS PNET cells showed an increased proportion of G1-phase cells. RHPS4-induced phenotypes were concomitant with telomerase inhibition, manifested in a telomere length-independent manner and not associated with activated c-Myc levels. However, anti-proliferative effects were also observed in normal neural/endothelial cells in vitro and ex vivo. CONCLUSION: This study warrants in vivo validation of RHPS4 and alternative G4 ligands as potential anti-cancer agents for brain tumors but highlights the consideration of dose-limiting tissue toxicities.


Subject(s)
Acridines/pharmacology , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Acridines/chemistry , Animals , Antineoplastic Agents/chemistry , Brain Neoplasms , Cell Line, Tumor , Cell Survival/drug effects , Cilia/drug effects , Cilia/physiology , Drug Screening Assays, Antitumor , Endothelial Cells/drug effects , Ependyma/pathology , G-Quadruplexes , Humans , Inhibitory Concentration 50 , Proto-Oncogene Proteins c-myc/metabolism , Rats , Taq Polymerase/antagonists & inhibitors , Telomerase/antagonists & inhibitors , Telomerase/chemistry , Telomere Homeostasis/drug effects
15.
Bioorg Med Chem ; 20(4): 1607-15, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22264753

ABSTRACT

Quinols have been developed as a class of potential anti-cancer compounds. They are thought to act as double Michael acceptors, forming two covalent bonds to their target protein(s). Quinols have also been shown to have activity against the parasite Trypanosoma brucei, the causative organism of human African trypanosomiasis, but they demonstrated little selectivity over mammalian MRC5 cells in a counter-screen. In this paper, we report screening of further examples of quinols against T. brucei. We were able to derive an SAR, but the compounds demonstrated little selectivity over MRC5 cells. In an approach to increase selectivity, we attached melamine and benzamidine motifs to the quinols, because these moieties are known to be selectively concentrated in the parasite by transporter proteins. In general these transporter motif-containing analogues showed increased selectivity; however they also showed reduced levels of potency against T. brucei.


Subject(s)
Drug Delivery Systems , Hydroquinones/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Benzamidines/chemical synthesis , Benzamidines/chemistry , Benzamidines/pharmacology , Cell Line , Humans , Hydroquinones/chemical synthesis , Hydroquinones/chemistry , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Triazines/chemical synthesis , Triazines/chemistry , Triazines/pharmacology , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/chemistry
16.
Curr Mol Pharmacol ; 5(1): 102-14, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22122467

ABSTRACT

Glioblastoma multiforme is the most common aggressive adult primary tumour of the central nervous system. Treatment includes surgery, radiotherapy and adjuvant temozolomide (TMZ) chemotherapy. TMZ is an alkylating agent prodrug, delivering a methyl group to purine bases of DNA (O6-guanine; N7-guanine and N3-adenine). The primary cytotoxic lesion, O6-methylguanine (O6-MeG) can be removed by methylguanine methyltransferase (MGMT; direct repair) in tumours expressing this protein, or tolerated in mismatch repair-deficient (MMR-) tumours. Thus MGMT or MMR deficiency confers resistance to TMZ. Inherent- and acquired resistance to TMZ present major obstacles to successful treatment. Strategies devised to thwart resistance and enhance response to TMZ, including inhibition of DNA repair mechanisms which contribute to TMZ resistance, are under clinical evaluation. Depletion of MGMT prior to alkylating agent chemotherapy prevents O6-MeG repair; thus, MGMT pseudosubstrates O6-benzylguanine and lomeguatrib are able to sensitise tumours to TMZ. Disruption of base excision repair (BER) results in persistence of potentially lethal N7- and N3- purine lesions contributing significantly to TMZ cytoxicity particularly when O6-MeG adducts are repaired or tolerated. Several small molecule inhibitors of poly(ADP-ribose)polymerase-1 (PARP-1), a critical BER protein are yielding promising results clinically, both in combination with TMZ and as single agent chemotherapy in patients whose tumours possess homologous recombination DNA repair defects. Another validated, but as yet preclinical protein target, mandatory to BER is abasic (AP) endonuclease-1 (APE-1); in preclinical tests, APE-1 inhibition potentiates TMZ activity. An alternative strategy is synthesis of a molecule, evoking an irrepairable cytotoxic O6-G lesion. Preliminary efforts to achieve this goal are described.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Repair/drug effects , Dacarbazine/analogs & derivatives , Antineoplastic Agents, Alkylating/therapeutic use , Cell Line, Tumor , Central Nervous System Neoplasms/drug therapy , DNA Glycosylases/metabolism , DNA Modification Methylases/chemistry , DNA Modification Methylases/metabolism , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Guanine/analogs & derivatives , Guanine/chemistry , Humans , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Temozolomide , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism
17.
Mol Cancer Ther ; 10(10): 1982-92, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21831963

ABSTRACT

Both 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (5F-203; NSC 703786) and 2-(3,4-dimethoxyphenyl)-5-fluorobenzothiazole (GW-610; NSC 721648) are antitumor agents with novel mechanism(s). Previous studies have indicated that cytochrome (CYP) P450 1A1 is crucial for 5F-203 activity. In the present study, we investigated the functional role of 2 newly identified CYP P450 enzymes, CYP2S1 and CYP2W1, in mediating antitumor activity of benzothiazole compounds. We generated isogenic breast cancer (MDA-MB-468, MCF-7) and colorectal cancer (CRC; KM12 and HCC2998) cell lines depleted for CYP1A1, CYP2S1, or CYP2W1. The sensitivity of these cells to 5F-203 and GW-610 was then compared with vector control cells. 5F-203 exhibited potent activity against breast cancer cells, whereas GW-610 was effective against both breast and colorectal cancer cells. CYP1A1 was induced in both breast cancer and CRC cells, while CYP2S1 and CYP2W1 were selectively induced in breast cancer cells only following treatment with 5F-203 or GW-610. Depletion of CYP1A1 abrogated the sensitivity of breast cancer and CRC cells to 5F-203 and GW-610. Although depletion of CYP2S1 sensitized both breast cancer and CRC cells toward 5F-203 and GW-610, CYP2W1 knockdown caused marked resistance to GW-610 in CRC cells. Our results indicate that CYP-P450 isoforms, with the exception of CYP1A1, play an important role in mediating benzothiazole activity. CYP2S1 appears to be involved in deactivation of benzothiazoles, whereas CYP2W1 is important for bioactivation of GW-610 in CRC cells. Because CYP2W1 is highly expressed in colorectal tumors, GW-610 represents a promising agent for CRC therapy.


Subject(s)
Benzothiazoles/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Cytochrome P-450 Enzyme System/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Colorectal Neoplasms/pathology , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P450 Family 2 , Enzyme Induction/drug effects , Female , Humans , Male , Thiazoles/pharmacology
18.
Oncology ; 80(3-4): 195-207, 2011.
Article in English | MEDLINE | ID: mdl-21720182

ABSTRACT

Resistance to temozolomide (TMZ), conferred by O6-methylguanine-DNA methyltransferase (MGMT) or mismatch repair (MMR) deficiency, presents obstacles to successful glioblastoma multiforme (GBM) treatment. Activities of novel TMZ analogs, designed to overcome resistance, were tested against isogenic SNB19 and U373 GBM cell lines (V = vector control, low MGMT; M = MGMT overexpression). TMZ and triazene MTIC demonstrated >9-fold resistance in SNB19M cells (cf SNB19V). N-3 methyl ester analog 11 and corresponding triazene 12 inhibited growth of TMZ-sensitive (V) and TMZ-resistant (M) cells (GI(50) <50 µM). Ethyl ester 13 and triazene 14 gave similar profiles. MMR-deficient colorectal carcinoma cells, resistant to TMZ (GI(50) >500 µM), responded to analog 11 and 13 treatment. Cross-resistance to these agents was not observed in cell lines possessing acquired TMZ resistance (SNB19VR; U373VR). Methyl ester 11 blocked SNB19V, SNB19M and SNB19VR cells in S and G(2)/M, causing dose- and time-dependent apoptosis. DNA damage, recruiting excision repair was detected by alkaline comet assay; H2AX phosphorylation indicated a lethal DNA double-strand break formation following analog 11 exposure. Compounds 11 and 13 demonstrated 3.7- and 5.1-fold enhanced activity in base excision repair-deficient Chinese hamster ovary cells; furthermore, poly (ADP-ribose) polymerase-1 inhibition potentiated HCT-116 cells' sensitivity to analog 11. In conclusion, analogs 11 and 13 exert anticancer activity irrespective of MGMT and MMR.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cell Cycle/drug effects , DNA Mismatch Repair/drug effects , DNA Repair/drug effects , DNA, Neoplasm/drug effects , Dacarbazine/analogs & derivatives , Glioma/genetics , Imidazoles/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/genetics , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Dacarbazine/pharmacology , Drug Resistance, Neoplasm/genetics , Glioma/drug therapy , Glioma/enzymology , Humans , Pilot Projects , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/drug effects , Promoter Regions, Genetic/drug effects , Temozolomide
19.
Clin Cancer Res ; 17(8): 2227-36, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21355072

ABSTRACT

PURPOSE: We previously reported that the G-quadruplex (G4) ligand RHPS4 potentiates the antitumor activity of camptothecins both in vitro and in tumor xenografts. The present study aims at investigating the mechanisms involved in this specific drug interaction. EXPERIMENTAL DESIGN: Combination index test was used to evaluate the interaction between G4 ligands and standard or novel Topo I inhibitors. Chromatin immunoprecipitation was performed to study the presence at telomeres of various types of topisomerase, while immunolabeling experiments were performed to measure the activation of DNA damage both in vitro and in tumor xenografts. RESULTS: We report that integration of the Topo I inhibitor SN-38, but not the Topo II poison doxorubicin with telomere-based therapy is strongly effective and the sequence of drug administration is critical in determining the synergistic interaction, impairing the cell ability to recover from drug-induced cytotoxicity. The synergistic effect of this combination was also observed by using novel camptothecins and, more interestingly, mice treated with ST1481/RHPS4 combination showed an inhibition and delay of tumor growth as well as an increased survival. The study of the mechanism(s) revealed that treatment with G4 ligands increased Topo I at the telomeres and the functional relevance of this observation was directly assessed by showing that standard and novel camptothecins stabilized DNA damage both in vitro and in xenografts. CONCLUSIONS: Our results demonstrate an outstanding efficacy of Topo I inhibitors/G4 ligands combination, which likely reflects an enhanced and persistent activation of DNA damage response as a critical determinant of the therapeutic improvement.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/drug therapy , DNA Damage , Xenograft Model Antitumor Assays , Acridines/administration & dosage , Acridines/pharmacology , Animals , Apoptosis/drug effects , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Cycle/drug effects , Cell Survival/drug effects , Cells, Cultured , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Synergism , G-Quadruplexes/drug effects , HCT116 Cells , HT29 Cells , Humans , Irinotecan , Male , Mice , Mice, Nude , Telomere/drug effects , Telomere/genetics , Telomere/metabolism , Topoisomerase I Inhibitors/administration & dosage , Topoisomerase I Inhibitors/pharmacology , Treatment Outcome
20.
Org Biomol Chem ; 8(9): 2078-84, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20401384

ABSTRACT

4-Ethynyl-4-hydroxycyclohexa-2,5-dien-1-one 5 undergoes cycloaddition reactions with a range of substituted azides in the presence of copper salts to form 1,4-disubstituted triazoles 8-11 bearing the 4-hydroxycyclohexa-2,5-dien-1-one (quinol) pharmacophore; one example of an isomeric 1,5-disubstituted triazole 12 was formed from 5 and benzyl azide in the presence of a ruthenium catalyst. Compounds were screened for growth-inhibitory activity against five cancer cell lines of colon, breast and lung origin, but were overall less potent than the benzothiazolyl- and indolyl-substituted quinols 2 and 3.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Copper/chemistry , Cyclohexanones/chemical synthesis , Cyclohexanones/pharmacology , Triazoles/chemical synthesis , Triazoles/pharmacology , Antineoplastic Agents/chemistry , Catalysis , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclization , Cyclohexanones/chemistry , Drug Screening Assays, Antitumor , Humans , Structure-Activity Relationship , Triazoles/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...