Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Anal Chem ; 94(22): 7804-7813, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35616489

RESUMO

Feature extraction algorithms are an important class of unsupervised methods used to reduce data dimensionality. They have been applied extensively for time-of-flight secondary ion mass spectrometry (ToF-SIMS) imaging─commonly, matrix factorization (MF) techniques such as principal component analysis have been used. A limitation of MF is the assumption of linearity, which is generally not accurate for ToF-SIMS data. Recently, nonlinear autoencoders have been shown to outperform MF techniques for ToF-SIMS image feature extraction. However, another limitation of most feature extraction methods (including autoencoders) that is particularly important for hyperspectral data is that they do not consider spatial information. To address this limitation, we describe the application of the convolutional autoencoder (CNNAE) to hyperspectral ToF-SIMS imaging data. The CNNAE is an artificial neural network developed specifically for hyperspectral data that uses convolutional layers for image encoding, thereby explicitly incorporating pixel neighborhood information. We compared the performance of the CNNAE with other common feature extraction algorithms for two biological ToF-SIMS imaging data sets. We investigated the extracted features and used the dimensionality-reduced data to train additional ML algorithms. By converting two-dimensional convolutional layers to three-dimensional (3D), we also showed how the CNNAE can be extended to 3D ToF-SIMS images. In general, the CNNAE produced features with significantly higher contrast and autocorrelation than other techniques. Furthermore, histologically recognizable features in the data were more accurately represented. The extension of the CNNAE to 3D data also provided an important proof of principle for the analysis of more complex 3D data sets.


Assuntos
Redes Neurais de Computação , Espectrometria de Massa de Íon Secundário , Algoritmos , Análise de Componente Principal , Espectrometria de Massa de Íon Secundário/métodos
2.
Cancer Chemother Pharmacol ; 89(6): 773-784, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35460360

RESUMO

PURPOSE: Pixantrone is a synthetic aza-anthracenedione currently used in the treatment of non-Hodgkin's lymphoma. The drug is firmly established as a poison of the nuclear enzyme topoisomerase II, however, pixantrone can also generate covalent drug-DNA adducts following activation by formaldehyde. While pixantrone-DNA adducts form proficiently in vitro, little evidence is presently at hand to indicate their existence within cells. The molecular nature of these lesions within cancer cells exposed to pixantrone and formaldehyde-releasing prodrugs was characterized along with the cellular responses to their formation. METHODS: In vitro crosslinking assays, [14C] scintillation counting analyses and alkaline comet assays were applied to characterize pixantrone-DNA adducts. Flow cytometry, cell growth inhibition and clonogenic assays were used to measure cancer cell kill and survival. RESULTS: Pixantrone-DNA adducts were not detectable in MCF-7 breast cancer cells exposed to [14C] pixantrone (10-40 µM) alone, however the addition of the formaldehyde-releasing prodrug AN9 yielded readily measurable levels of the lesion at ~ 1 adduct per 10 kb of genomic DNA. Co-administration with AN9 completely reversed topoisomerase II-associated DNA damage induction by pixantrone yet potentiated cell kill by the drug, suggesting that pixantrone-DNA adducts may promote a topoisomerase II-independent mechanism of cell death. Pixantrone-DNA adduct-forming treatments generally conferred mild synergism in multiple cell lines in various cell death and clonogenic assays, while pixantrone analogues either incapable or relatively defective in forming DNA adducts demonstrated antagonism when combined with AN9. CONCLUSIONS: The features unique to pixantrone-DNA adducts may be leveraged to enhance cancer cell kill and may be used to guide the design of pixantrone analogues that generate adducts with more favorable anticancer properties.


Assuntos
Neoplasias , Pró-Fármacos , Adutos de DNA , DNA Topoisomerases Tipo II/metabolismo , Formaldeído/farmacologia , Humanos , Isoquinolinas , Pró-Fármacos/farmacologia
3.
Lab Anim ; 55(4): 367-374, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33874818

RESUMO

NSG™ mice are highly immunocompromised thus demonstrate high efficiency engraftment of patient-derived xenografts (PDXs) for pre-clinical oncology research. It has previously been reported that NSG™ mice are hyper-sensitive to doxorubicin due to the impairment of DNA damage repair mechanisms. As such, doxorubicin causes a wide spectrum of toxicities including cardiotoxicity, hepatotoxicity and intestinal toxicity in NSG™ mice. Doxil is an alternative clinical formulation of doxorubicin, where doxorubicin is encapsulated within pegylated liposomes and displays improved toxicity profiles compared to conventional doxorubicin. Doxil was substituted for doxorubicin in our study to determine its toxicity profile in female NSG™ mice. The mice that were treated with Doxil developed dose-dependent histopathological alterations associated with non-glandular gastritis, with non-Helicobacter spp. bacterial infiltrates, as well as oesophagitis. Of note, a study using a dose of 2 mg/kg Doxil was terminated early due to significant weight loss while the use of Doxil at 1 mg/kg allowed for repeated treatment of twice a week for a duration of three weeks. A dose optimised treatment regimen has now been established and can be applied to assess Doxil-related anti-tumour efficacy in a range of PDX-bearing NSG™ mice.


Assuntos
Doxorrubicina , Gastrite , Animais , Doxorrubicina/análogos & derivados , Doxorrubicina/toxicidade , Feminino , Gastrite/induzido quimicamente , Lipossomos , Camundongos , Polietilenoglicóis
4.
Biochem Pharmacol ; 185: 114410, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33428897

RESUMO

Cancer patients treated with doxorubicin are at risk of congestive heart failure due to doxorubicin-mediated cardiotoxicity via topoisomerase IIß poisoning. Acute cardiac muscle damage occurs in response to the very first dose of doxorubicin, however, cardioprotection has been reported after co-treatment of doxorubicin with acyloxyalkyl ester prodrugs. The aim of this study was to examine the role played by various forms of acute cardiac damage mediated by doxorubicin and determine a mechanism for the cardioprotective effect of formaldehyde-releasing prodrug AN-9 (pivaloyloxymethyl butyrate). Doxorubicin-induced cardiac damage in BALB/c mice bearing mammary tumours was established with a single dose of doxorubicin (4 or 16 mg/kg) administered alone or in combination with AN-9 (100 mg/kg). AN-9 protected the heart from doxorubicin-induced myocardial apoptosis and also significantly reduced dsDNA breaks, independent from the level of doxorubicin biodistribution to the heart. Covalent incorporation of [14C]doxorubicin into DNA showed that the combination treatment yielded significantly higher levels of formaldehyde-mediated doxorubicin-DNA adducts compared to doxorubicin alone, yet this form of damage was associated with cardioprotection from apoptosis. The cardiac transcriptomic analysis indicates that the combination treatment initiates inflammatory response signalling pathways. Doxorubicin and AN-9 combination treatments were cardioprotective, yet preserved doxorubicin-mediated anti-tumour proliferation and apoptosis in mammary tumours. This was associated with a switch in doxorubicin action from cardiac topoisomerase IIß poisoning to covalent-DNA adduct formation. Co-administration of doxorubicin and formaldehyde-releasing prodrugs, such as AN-9, may be a promising cardioprotective therapy while maintaining doxorubicin activity in primary mammary tumours.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Cardiotoxicidade/patologia , Cardiotoxicidade/prevenção & controle , Doxorrubicina/toxicidade , Miocárdio/patologia , Animais , Cardiotônicos/farmacologia , Cardiotônicos/uso terapêutico , Cardiotoxicidade/metabolismo , Relação Dose-Resposta a Droga , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Miocárdio/metabolismo
5.
Bioorg Med Chem Lett ; 35: 127813, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33486050

RESUMO

Current techniques for the identification of DNA adduct-inducing and DNA interstrand crosslinking agents include electrophoretic crosslinking assays, electrophoretic gel shift assays, DNA and RNA stop assays, mass spectrometry-based methods and 32P-post-labelling. While these assays provide considerable insight into the site and stability of the interaction, they are relatively expensive, time-consuming and sometimes rely on the use of radioactively-labelled components, and thus are ill-suited to screening large numbers of compounds. A novel medium throughput assay was developed to overcome these limitations and was based on the attachment of a biotin-tagged double stranded (ds) oligonucleotide to Corning DNA-Bind plates. We aimed to detect anthracycline and anthracenedione DNA adducts which form by initial non-covalent intercalation with duplex DNA, and subsequent covalent adduct formation which is mediated by formaldehyde. Following drug treatment, DNA samples were subjected to a denaturation step, washing and then measurement by fluorescence to detect remaining drug-DNA species using streptavidin-europium. This dissociation-enhanced lanthanide fluorescent immunoassay (DELFIA) is a time-resolved fluorescence intensity assay where the fluorescence signal arises only from stabilised drug-DNA complexes. We applied this new methodology to the identification of anthracycline-like compounds with the ability to functionally crosslink double-strand oligonucleotides. The entire procedure can be performed by robotics, requiring low volumes of compounds and reagents, thereby reducing costs and enabling multiple compounds to be assessed on a single microtitre plate.


Assuntos
Automação , Reagentes de Ligações Cruzadas/farmacologia , Adutos de DNA/efeitos dos fármacos , Desenvolvimento de Medicamentos , Reagentes de Ligações Cruzadas/síntese química , Reagentes de Ligações Cruzadas/química , Relação Dose-Resposta a Droga , Estrutura Molecular , Relação Estrutura-Atividade
6.
Biopolymers ; 112(4): e23400, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32937683

RESUMO

The application of artificial intelligence and machine learning to hyperspectral mass spectrometry imaging (MSI) data has received considerable attention over recent years. Various methodologies have shown great promise in their ability to handle the complexity and size of MSI data sets. Advances in this area have been particularly appealing for MSI of biological samples, which typically produce highly complicated data with often subtle relationships between features. There are many different machine learning approaches that have been applied to MSI data over the past two decades. In this review, we focus on a subset of non-linear machine learning techniques that have mostly only been applied in the past 5 years. Specifically, we review the use of the self-organizing map (SOM), SOM with relational perspective mapping (SOM-RPM), t-distributed stochastic neighbor embedding (t-SNE) and uniform manifold approximation and projection (UMAP). While not their only functionality, we have grouped these techniques based on their ability to produce what we refer to as similarity maps. Similarity maps are color representations of hyperspectral data, in which spectral similarity between pixels-that is, their distance in high-dimensional space-is represented by relative color similarity. In discussing these techniques, we describe, briefly, their associated algorithms and functionalities, and also outline applications in MSI research with a strong focus on biological sample types. The aim of this review is therefore to introduce this relatively recent paradigm for visualizing and exploring hyperspectral MSI, while also providing a comparison between each technique discussed.


Assuntos
Imageamento Hiperespectral/métodos , Aprendizado de Máquina , Espectrometria de Massas/métodos , Algoritmos , Animais , Inteligência Artificial , Humanos
7.
Mol Neurodegener ; 15(1): 51, 2020 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-32907630

RESUMO

BACKGROUND: Pathological forms of TAR DNA-binding protein 43 (TDP-43) are present in motor neurons of almost all amyotrophic lateral sclerosis (ALS) patients, and mutations in TDP-43 are also present in ALS. Loss and gain of TDP-43 functions are implicated in pathogenesis, but the mechanisms are unclear. While the RNA functions of TDP-43 have been widely investigated, its DNA binding roles remain unclear. However, recent studies have implicated a role for TDP-43 in the DNA damage response. METHODS: We used NSC-34 motor neuron-like cells and primary cortical neurons expressing wildtype TDP-43 or TDP-43 ALS associated mutants (A315T, Q331K), in which DNA damage was induced by etoposide or H2O2 treatment. We investigated the consequences of depletion of TDP-43 on DNA repair using small interfering RNAs. Specific non homologous end joining (NHEJ) reporters (EJ5GFP and EJ2GFP) and cells lacking DNA-dependent serine/threonine protein kinase (DNA-PK) were used to investigate the role of TDP-43 in DNA repair. To investigate the recruitment of TDP-43 to sites of DNA damage we used single molecule super-resolution microscopy and a co-immunoprecipitation assay. We also investigated DNA damage in an ALS transgenic mouse model, in which TDP-43 accumulates pathologically in the cytoplasm. We also examined fibroblasts derived from ALS patients bearing the TDP-43 M337V mutation for evidence of DNA damage. RESULTS: We demonstrate that wildtype TDP-43 is recruited to sites of DNA damage where it participates in classical NHEJ DNA repair. However, ALS-associated TDP-43 mutants lose this activity, which induces DNA damage. Furthermore, DNA damage is present in mice displaying TDP-43 pathology, implying an active role in neurodegeneration. Additionally, DNA damage triggers features typical of TDP-43 pathology; cytoplasmic mis-localisation and stress granule formation. Similarly, inhibition of NHEJ induces TDP-43 mis-localisation to the cytoplasm. CONCLUSIONS: This study reveals that TDP-43 functions in DNA repair, but loss of this function triggers DNA damage and is associated with key pathological features of ALS.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Dano ao DNA/fisiologia , Reparo do DNA por Junção de Extremidades/fisiologia , Proteínas de Ligação a DNA/metabolismo , Adulto , Idoso , Animais , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Neurônios Motores/metabolismo
8.
Anal Chem ; 92(15): 10450-10459, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32614172

RESUMO

We present an optimization of the toroidal self-organizing map (SOM) algorithm for the accurate visualization of hyperspectral data. This represents a significant advancement on our previous work, in which we demonstrated the use of toroidal SOMs for the visualization of time-of-flight secondary ion mass spectrometry (ToF-SIMS) imaging data. We have previously shown that the toroidal SOM can be used, unsupervised, to produce a multicolor similarity map of the analysis area, in which pixels with similar mass spectra are assigned a similar color. Here, we use an additional algorithm, relational perspective mapping (RPM), to produce more accurate visualizations of hyperspectral data. The SOM output is used as an input for the RPM algorithm, which is a nonlinear dimensionality reduction technique designed to produce a two-dimensional map of high-dimensional data. Using the topological information provided by the SOM, RPM provides complementary distance information. The result is a color scheme that more accurately reflects the local spectral distances between pixels in the data. We exemplify SOM-RPM using ToF-SIMS imaging data from a mouse tumor tissue section. The similarity maps produced are compared with those produced by two leading hyperspectral visualization techniques in the field of mass spectrometry imaging: t-distributed stochastic neighborhood embedding (t-SNE) and uniform manifold approximation and projection (UMAP). We evaluate the performance of each technique both qualitatively and quantitatively, investigating the correlations between distances in the models and distances in the data. SOM-RPM is demonstrably highly competitive with t-SNE and UMAP, according to our evaluations. Furthermore, the use of a neural network offers distinct advantages in data characterization, which we discuss. We also show how spectra extracted from regions of interest identified by SOM-RPM can be further analyzed using linear discriminant analysis for the validation and characterization of the surface chemistry.

9.
Anal Chem ; 92(9): 6587-6597, 2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32233419

RESUMO

Combinatorial approaches to materials discovery offer promising potential for the rapid development of novel polymer systems. Polymer microarrays enable the high-throughput comparison of material physical and chemical properties-such as surface chemistry and properties like cell attachment or protein adsorption-in order to identify correlations that can progress materials development. A challenge for this approach is to accurately discriminate between highly similar polymer chemistries or identify heterogeneities within individual polymer spots. Time-of-flight secondary ion mass spectrometry (ToF-SIMS) offers unique potential in this regard, capable of describing the chemistry associated with the outermost layer of a sample with high spatial resolution and chemical sensitivity. However, this comes at the cost of generating large scale, complex hyperspectral imaging data sets. We have demonstrated previously that machine learning is a powerful tool for interpreting ToF-SIMS images, describing a method for color-tagging the output of a self-organizing map (SOM). This reduces the entire hyperspectral data set to a single reconstructed color similarity map, in which the spectral similarity between pixels is represented by color similarity in the map. Here, we apply the same methodology to a ToF-SIMS image of a printed polymer microarray for the first time. We report complete, single-pixel molecular discrimination of the 70 unique homopolymer spots on the array while also identifying intraspot heterogeneities thought to be related to intermixing of the polymer and the pHEMA coating. In this way, we show that the SOM can identify layers of similarity and clusters in the data, both with respect to polymer backbone structures and their individual side groups. Finally, we relate the output of the SOM analysis with fluorescence data from polymer-protein adsorption studies, highlighting how polymer performance can be visualized within the context of the global topology of the data set.

10.
Bioorg Med Chem ; 28(3): 115260, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31870833

RESUMO

Mitoxantrone is an anticancer anthracenedione that can be activated by formaldehyde to generate covalent drug-DNA adducts. Despite their covalent nature, these DNA lesions are relatively labile. It was recently established that analogues of mitoxantrone featuring extended side-chains terminating in primary amino groups typically yielded high levels of stable DNA adducts following their activation by formaldehyde. In this study we describe the DNA sequence-specific binding properties of the mitoxantrone analogue WEHI-150 which is the first anthracenedione to form apparent DNA crosslinks mediated by formaldehyde. The utility of this compound lies in the versatility of the covalent binding modes displayed. Unlike other anthracenediones described to date, WEHI-150 can mediate covalent adducts that are independent of interactions with the N-2 of guanine and is capable of adduct formation at novel DNA sequences. Moreover, these covalent adducts incorporate more than one formaldehyde-mediated bond with DNA, thus facilitating the formation of highly lethal DNA crosslinks. The versatility of binding observed is anticipated to allow the next generation of anthracenediones to interact with a broader spectrum of nucleic acid species than previously demonstrated by the parent compounds, thus allowing for more diverse biological activities.


Assuntos
DNA/efeitos dos fármacos , Formaldeído/farmacologia , Mitoxantrona/farmacologia , Animais , Bovinos , Relação Dose-Resposta a Droga , Formaldeído/química , Espectrometria de Massas , Mitoxantrona/análogos & derivados , Mitoxantrona/química , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade
11.
Anal Chem ; 91(21): 13855-13865, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31549810

RESUMO

Time-of-flight secondary ion mass spectrometry (ToF-SIMS) is a powerful surface characterization technique capable of producing high spatial resolution hyperspectral images, in which each pixel comprises an entire mass spectrum. Such images can provide insight into the chemical composition across a surface. However, issues arise due to the size and complexity of the data produced. Data are particularly complicated for biological samples, primarily due to overlapping spectra produced by similar components. The traditional approach of selecting individual ion peaks as representative of particular components is insufficient for such complex data sets. Multivariate analysis (MVA) can help to overcome this significant hurdle. We demonstrate that Kohonen self-organizing maps (SOMs) with a toroidal topology can be used to analyze a ToF-SIMS hyperspectral imaging data set and identify spectral similarities between pixels. We present a method for color-tagging the toroidal SOM output, which reduces the entire data set to a single RGB image in which similar pixels-based on their associated mass spectra-are assigned a similar color. This method was exemplified using a ToF-SIMS image of dried large multilamellar vesicles (LMVs), loaded with the antibiotic cefditoren pivoxil (CP). We successfully identified CP-loaded and empty LMVs without the need for any prior knowledge of the sample, despite their highly similar spectra. We also identified which specific ion peaks were most important in differentiating the two LMV populations. This approach is entirely unsupervised and requires minimal experimenter input. It was developed with the aim of providing a user-friendly yet sophisticated workflow for understanding complex biological samples using ToF-SIMS images.

12.
ACS Med Chem Lett ; 8(5): 538-542, 2017 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-28523107

RESUMO

Mitoxantrone was efficiently encapsulated within cucurbit[8]uril in a 2:1 complex where the two mitoxantrone molecules were symmetrically located through both portals of a cucurbit[8]uril cage. The novel complex facilitates increased mitoxantrone uptake in mouse breast cancer cells and decreases the toxicity of the drug in healthy mice. In an orthotopic mouse model of metastatic breast cancer the complex still maintains in vivo anticancer activity compared to the free drug, yet provides a statistically significant increase in the survival of these mice compared to conventional mitoxantrone treatment. This new low toxicity formulation offers the possibility to increase mitoxantrone dose and thus maximize efficacy while managing the dose limiting side effects.

13.
Invest New Drugs ; 35(4): 412-426, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28315153

RESUMO

The histone deacetylase (HDAC) inhibitory prodrugs of butyric (AN7) and valproic (AN446) acids, which release the active acids upon metabolic degradation, were studied examining their differential effects on the viability, HDAC inhibitory activity and the DNA damage response (DDR), in glioblastoma cell and normal human astrocytes (NHAs). In xenografts of glioblastoma, AN7 or AN446 given or the combination of each of them with Dox augmented the anticancer activity of Dox and protected the heart from its toxicity. In order to determine the processes underlying these opposing effects, the changes induced by these treatments on the epigenetic landscape, the DDR, and fibrosis were compared in tumors and hearts of glioblastoma xenografts. The potency of AN7 and AN446 as HDAC inhibitors was correlated with their effects on the viability of the cancer and non-cancer cells. The prodrugs affected the epigenetic landscape and the DDR in a tissue-specific and context-dependent manner. Findings suggest that the selectivity of the prodrugs could be attributed to their different effects on histone modification patterns in normal vs. transformed tissues. Further studies are warranted to substantiate the potential of AN446 as a new anticancer drug for glioblastoma patients.


Assuntos
Antineoplásicos/farmacologia , Epigênese Genética/efeitos dos fármacos , Glioblastoma/genética , Inibidores de Histona Desacetilases/farmacologia , Pró-Fármacos/farmacologia , Acetilação/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Encéfalo/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA , Doxorrubicina/farmacologia , Doxorrubicina/toxicidade , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Inibidores de Histona Desacetilases/uso terapêutico , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Metilação/efeitos dos fármacos , Camundongos Nus , Miocárdio/metabolismo , Pró-Fármacos/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Org Biomol Chem ; 14(43): 10217-10221, 2016 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-27735959

RESUMO

The major covalent adduct formed between a 13C-labelled formaldehyde activated bis-amino mitoxantrone analogue (WEHI-150) and the hexanucleotide d(CG5MeCGCG)2 has been isolated by HPLC chromatography and the structure determined by NMR spectroscopy. The results indicate that WEHI-150 forms one covalent bond through a primary amine to the N-2 of the G2 residue, with the polycyclic ring structure intercalated at the 5MeC3pG4/G10p5MeC9 site. Furthermore, the WEHI-150 aromatic ring system is oriented approximately parallel to the long axis of the base pairs, with one aliphatic side-chain in the major groove and the other side-chain in the minor groove. This study indicates that mitoxantrone derivatives like WEHI-150 should be capable of forming major-minor groove cross-linked adducts that will likely produce considerably different intracellular biological properties compared to known anthracycline and anthracenedione anticancer drugs.


Assuntos
DNA/química , Mitoxantrona/química , Conformação de Ácido Nucleico , Modelos Moleculares , Oligonucleotídeos/química
15.
Org Biomol Chem ; 14(20): 4728-38, 2016 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-27142235

RESUMO

The ability of a bis-amino mitoxantrone anticancer drug (named WEHI-150) to form covalent adducts with DNA, after activation by formaldehyde, has been studied by electrospray ionisation mass spectrometry and HPLC. Mass spectrometry results showed that WEHI-150 could form covalent adducts with d(ACGCGCGT)2 that contained one, two or three covalent links to the octanucleotide, whereas the control drugs (daunorubicin and the anthracenediones mitoxantrone and pixantrone) only formed adducts with one covalent link to the octanucleotide. HPLC was used to examine the extent of covalent bond formation of WEHI-150 with d(CGCGCG)2 and d(CG(5Me)CGCG)2. Incubation of WEHI-150 with d(CG(5Me)CGCG)2 in the presence of formaldehyde resulted in the formation of significantly greater amounts of covalent adducts than was observed with d(CGCGCG)2. In order to understand the observed increase of covalent adducts with d(CG(5Me)CGCG)2, an NMR study of the reversible interaction of WEHI-150 at both CpG and (5Me)CpG sites was undertaken. Intermolecular NOEs were observed in the NOESY spectra of d(ACGGCCGT)2 with added WEHI-150 that indicated that the drug selectively intercalated at the CpG sites and from the major groove. In particular, NOEs were observed from the WEHI-150 H2,3 protons to the H1' protons of G3 and G7 and from the H6,7 protons to the H5 protons of C2 and C6. By contrast, intermolecular NOEs were observed between the WEHI-150 H2,3 protons to the H2'' proton of the (5Me)C3 in d(CG(5Me)CGCG)2, and between the drug aliphatic protons and the H1' proton of G4. This demonstrated that WEHI-150 preferentially intercalates at (5Me)CpG sites, compared to CpG sequences, and predominantly via the minor groove at the (5Me)CpG site. The results of this study demonstrate that WEHI-150 is likely to form interstrand DNA cross-links, upon activation by formaldehyde, and consequently exhibit greater cytotoxicity than other current anthracenedione drugs.


Assuntos
DNA/química , Formaldeído/química , Mitoxantrona/química , Sequência de Bases , Catálise , DNA/genética , Modelos Moleculares , Conformação de Ácido Nucleico
16.
Med Res Rev ; 36(2): 248-99, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26286294

RESUMO

Mitoxantrone is a synthetic anthracenedione originally developed to improve the therapeutic profile of the anthracyclines and is commonly applied in the treatment of breast and prostate cancers, lymphomas, and leukemias. A comprehensive overview of the drug's molecular, biochemical, and cellular pharmacology is presented here, beginning with the cardiotoxic nature of its predecessor doxorubicin and how these properties shaped the pharmacology of mitoxantrone itself. Although mitoxantrone is firmly established as a DNA topoisomerase II poison within mammalian cells, it is now clear that the drug interacts with a much broader range of biological macromolecules both covalently and noncovalently. Here, we consider each of these interactions in the context of their wider biological relevance to cancer therapy and highlight how they may be exploited to further enhance the therapeutic value of mitoxantrone. In doing so, it is now clear that mitoxantrone is more than just another topoisomerase II poison.


Assuntos
Mitoxantrona/farmacologia , Inibidores da Topoisomerase II/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Ensaios Clínicos como Assunto , Descoberta de Drogas , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Mitoxantrona/administração & dosagem , Mitoxantrona/química , Mitoxantrona/farmacocinética , Inibidores da Topoisomerase II/química
17.
Nanotechnol Sci Appl ; 8: 67-80, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26715842

RESUMO

Development of parameters for the fabrication of nanosized vectors is pivotal for its successful administration in therapeutic applications. In this study, homogeneously distributed chitosan nanoparticles (CNPs) with diameters as small as 62 nm and a polydispersity index (PDI) of 0.15 were synthesized and purified using a simple, robust method that was highly reproducible. Nanoparticles were synthesized using modified ionic gelation of the chitosan polymer with sodium tripolyphosphate. Using this method, larger aggregates were mechanically isolated from single particles in the nanoparticle population by selective efficient centrifugation. The presence of disaggregated monodisperse nanoparticles was confirmed using atomic force microscopy. Factors such as anions, pH, and concentration were found to affect the size and stability of nanoparticles directly. The smallest nanoparticle population was ∼62 nm in hydrodynamic size, with a low PDI of 0.15, indicating high particle homogeneity. CNPs were highly stable and retained their monodisperse morphology in serum-supplemented media in cell culture conditions for up to 72 hours, before slowly degrading over 6 days. Cell viability assays demonstrated that cells remained viable following a 72-hour exposure to 1 mg/mL CNPs, suggesting that the nanoparticles are well tolerated and highly suited for biomedical applications. Cellular uptake studies using fluorescein isothiocyanate-labeled CNPs showed that cancer cells readily accumulate the nanoparticles 30 minutes posttreatment and that nanoparticles persisted within cells for up to 24 hours posttreatment. As a proof of principle for use in anticancer therapeutic applications, a [(14)C]-radiolabeled form of the anticancer agent doxorubicin was efficiently encapsulated within the CNP, confirming the feasibility of using this system as a drug delivery vector.

18.
Org Biomol Chem ; 13(21): 5972-82, 2015 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-25929194

RESUMO

The binding of the anti-cancer drug pixantrone to three oligonucleotide sequences, d(TCATATGA)2, d(CCGAGAATTCCGG)2 {double bulge = DB} and the non-self complementary d(TACGATGAGTA) : d(TACCATCGTA) {single bulge = SB}, has been studied by NMR spectroscopy and molecular modelling. The upfield shifts observed for the aromatic resonances of pixantrone upon addition of the drug to each oligonucleotide confirmed the drug bound by intercalation. For the duplex sequence d(TCATATGA)2, NOEs were observed from the pixantrone aromatic H7/8 and aliphatic Ha/Hb protons to the H6/H8 and H1' protons of the C2, A3, T6 and G7 nucleotides, demonstrating that pixantrone preferentially binds at the symmetric CpA sites. However, weaker NOEs observed to various protons from the T4 and A5 residues indicated alternative minor binding sites. NOEs from the H7/H8 and Ha/Hb protons to both major (H6/H8) and minor groove (H1') protons indicated approximately equal proportions of intercalation was from the major and minor groove at the CpA sites. Intermolecular NOEs were observed between the H7/H8 and H4 protons of pixantrone and the A4H1' and G3H1' protons of the oligonucleotide that contains two symmetrically related bulge sites (DB), indicative of binding at the adenine bulge sites. For the oligonucleotide that only contains a single bulge site (SB), NOEs were observed from pixantrone protons to the SB G7H1', A8H1' and G9H1' protons, confirming that the drug bound selectively at the adenine bulge site. A molecular model of pixantrone-bound SB could be constructed with the drug bound from the minor groove at the A8pG9 site that was consistent with the observed NMR data. The results demonstrate that pixantrone preferentially intercalates at adenine bulge sites, compared to duplex DNA, and predominantly from the minor groove.


Assuntos
Antineoplásicos/farmacologia , DNA/química , Substâncias Intercalantes/farmacologia , Isoquinolinas/farmacologia , Sequência de Bases , Sítios de Ligação , Simulação de Acoplamento Molecular , Conformação de Ácido Nucleico
19.
Curr Top Med Chem ; 15(14): 1409-22, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25866273

RESUMO

Doxorubicin has been in use as a key anticancer drug for forty years, either as a single agent or in combination chemotherapy. It functions primarily by interfering with topoisomerase II activity but in the presence of formaldehyde, it forms adducts with DNA, mainly with the exocyclic amine of guanine at GpC sites and these adducts are more cytotoxic than topoisomerase II induced damage. High levels of adducts form spontaneously from the endogenous level of formaldehyde in tumour cells (1,300 adducts per cell after a 4 hr treatment with doxorubicin), but substantially higher levels form with the addition of exogenous sources of formaldehyde, such as formaldehyde releasing prodrugs. The enhanced cytotoxicity of adducts has been confirmed in mouse models, with adduct-forming conditions resulting in much improved inhibition of tumour growth, as well as cardioprotection. Doxorubicin cardiotoxicity has been attributed to topoisomerase II poisoning, and the cardioprotection is consistent with a mechanism switch from topoisomerase II poisoning to covalent adduct formation. Although the adducts have a half-life of less than one day, a population remains as essentially permanent lesions. The capacity of doxorubicin to form adducts offers a range of potential advantages over the conventional use of doxorubicin (as a topoisomerase II poison), including: enhanced cell kill; tumour-selective activation, hence tumour-selective cell kill; decreased cardiotoxicity; decreased resistance to prolonged doxorubicin treatment. There is therefore enormous potential to improve clinical responses to doxorubicin by using conditions which favour the formation of doxorubicin-DNA adducts.


Assuntos
Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacologia , Adutos de DNA/efeitos dos fármacos , Doxorrubicina/química , Doxorrubicina/farmacologia , Formaldeído/farmacologia , Adutos de DNA/química , Formaldeído/química , Humanos
20.
Mutat Res ; 777: 23-32, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25916945

RESUMO

Chemotherapy and radiotherapy can cause permanent damage to the genomes of surviving cells, provoking severe side effects such as second malignancies in some cancer survivors. Drugs that mimic the activity of death ligands, or antagonise pro-survival proteins of the Bcl-2 or IAP families have yielded encouraging results in animal experiments and early phase clinical trials. Because these agents directly engage apoptosis pathways, rather than damaging DNA to indirectly provoke tumour cell death, we reasoned that they may offer another important advantage over conventional therapies: minimisation or elimination of side effects such as second cancers that result from mutation of surviving normal cells. Disappointingly, however, we previously found that concentrations of death receptor agonists like TRAIL that would be present in vivo in clinical settings provoked DNA damage in surviving cells. In this study, we used cell line model systems to investigate the mutagenic capacity of drugs from two other classes of direct apoptosis-inducing agents: the BH3-mimetic ABT-737 and the IAP antagonists LCL161 and AT-406. Encouragingly, our data suggest that IAP antagonists possess negligible genotoxic activity. Doses of ABT-737 that were required to damage DNA stimulated Bax/Bak-independent signalling and exceeded concentrations detected in the plasma of animals treated with this drug. These findings provide hope that cancer patients treated by BH3-mimetics or IAP antagonists may avoid mutation-related illnesses that afflict some cancer survivors treated with conventional DNA-damaging anti-cancer therapies.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Proteínas Inibidoras de Apoptose/genética , Mutação , Proteína X Associada a bcl-2/genética , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Azocinas/farmacologia , Compostos Benzidrílicos/farmacologia , Compostos de Bifenilo/farmacologia , Linhagem Celular Tumoral , Ensaio Cometa , Dano ao DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Proteínas Inibidoras de Apoptose/metabolismo , Camundongos , Mutagênicos/farmacologia , Nitrofenóis/farmacologia , Piperazinas/farmacologia , Sulfonamidas/farmacologia , Tiazóis/farmacologia , Proteína X Associada a bcl-2/antagonistas & inibidores , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...