Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
BMC Cancer ; 13: 168, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23548101

RESUMO

BACKGROUND: Belinostat is a histone deacetylase inhibitor with anti-tumor effect in several pre-clinical tumor models and clinical trials. The aim of the study was to evaluate changes in cell proliferation and glucose uptake by use of 3'-deoxy-3'-[(18)F]fluorothymidine ([18F]FLT) and 2-deoxy-2-[(18)F]fluoro-D-glucose ([18F]FDG) positron emission tomography (PET) following treatment with belinostat in ovarian cancer in vivo models. METHODS: In vivo uptake of [18F]FLT and [18F]FDG in human ovary cancer xenografts in mice (A2780) were studied after treatment with belinostat. Mice were divided in 2 groups receiving either belinostat (40 mg/kg ip twice daily Day 0-4 and 6-10) or vehicle. Baseline [18F]FLT or [18F]FDG scans were made before treatment (Day 0) and repeated at Day 3, 6 and 10. Tracer uptake was quantified using small animal PET/CT. RESULTS: Tumors in the belinostat group had volumes that were 462 ± 62% (640 mm(3)) at Day 10 relative to baseline which was significantly different (P = 0.011) from the control group 769 ± 74% (926 mm(3)). [18F]FLT SUVmax increased from baseline to Day 10 (+30 ± 9%; P = 0.048) in the control group. No increase was observed in the treatment group. [18F]FDG SUVmean was significantly different in the treatment group compared to the control group (P = 0.0023) at Day 10. Within treatment groups [18F]FDG uptake and to a lesser extent [18F]FLT uptake at Day 3 were significantly correlated with tumor growth at Day 10. CONCLUSIONS: [18F]FDG uptake early following treatment initiation predicted tumor sizes at Day 10, suggesting that [18F]FDG may be a valuable biomarker for non-invasive assessment of anti-tumor activity of belinostat.


Assuntos
Didesoxinucleosídeos , Fluordesoxiglucose F18 , Neoplasias Ovarianas/diagnóstico , Tomografia por Emissão de Pósitrons , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Modelos Animais de Doenças , Feminino , Transportador de Glucose Tipo 1/genética , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Ácidos Hidroxâmicos/uso terapêutico , Antígeno Ki-67/genética , Camundongos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Compostos Radiofarmacêuticos , Sulfonamidas/administração & dosagem , Sulfonamidas/uso terapêutico , Timidina Quinase/genética , Carga Tumoral
2.
J Gene Med ; 14(7): 445-58, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22576955

RESUMO

BACKGROUND: Transcriptional targeted suicide gene (SG) therapy driven by the insulinoma-associated 1 (INSM1) promoter makes it possible to target suicide toxin production and cytotoxicity exclusively to small cell lung cancer (SCLC) cells and tumors. It remains to be determined whether acquired chemoresistance, as observed in the majority of SCLC patients, desensitizes SCLC cells to INSM1 promoter-driven SG therapy. METHODS: A panel of SCLC cell lines resistant to clinically relevant chemotherapeutics was characterized regarding the expression of proteins involved in response to chemotherapy and regarding INSM1 promoter activity. Sensitivity towards INSM1 promoter-driven SG therapy was tested using different systems: Yeast cytosine deaminase-uracil phosphoribosyl transferase (YCD-YUPRT) in combination with the prodrug 5-fluorocytosine (5-FC) or Escherichia coli nitroreductase (NTR) together with the bromomustard prodrug SN27686. RESULTS: The chemoresistant cell lines displayed heterogeneous expression profiles of molecules involved in multidrug resistance, apoptosis and survival pathways. Despite this, the INSM1 promoter activity was found to be unchanged or increased in SCLC chemoresistant cells and xenografts compared to chemosensitive variants. INSM1 promoter-driven SG therapy with YCD-YUPRT/5-FC or NTR/SN27686, was found to induce high levels of cytotoxicity in both chemosensitive and chemoresistant SCLC cells. Moreover, the combination of INSM1 promoter-driven YCD-YUPRT/5-FC therapy and chemotherapy, as well as the combination of INSM1 promoter-driven YCD-YUPRT/5-FC and NTR/SN27686 therapy, was observed to be superior to single agent therapy in chemoresistant SCLC cells. CONCLUSIONS: Collectively, the present study demonstrates that targeted SG therapy is a potent therapeutic approach for chemoresistant SCLC patients, with the highest efficacy achieved when applied as combination SG therapy or in combination with standard chemotherapy.


Assuntos
Genes Transgênicos Suicidas/genética , Terapia Genética/métodos , Neoplasias Pulmonares/terapia , Proteínas Repressoras/genética , Carcinoma de Pequenas Células do Pulmão/terapia , Animais , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , Quimioterapia Adjuvante , Citosina Desaminase/genética , Citosina Desaminase/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Quimioterapia Combinada , Escherichia coli/enzimologia , Escherichia coli/genética , Flucitosina/uso terapêutico , Humanos , Masculino , Camundongos , Nitrorredutases/uso terapêutico , Pentosiltransferases/genética , Pentosiltransferases/uso terapêutico , Regiões Promotoras Genéticas/genética , Leveduras/enzimologia , Leveduras/genética
3.
Nature ; 444(7119): 633-7, 2006 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-17136093

RESUMO

Recent studies have indicated the existence of tumorigenesis barriers that slow or inhibit the progression of preneoplastic lesions to neoplasia. One such barrier involves DNA replication stress, which leads to activation of the DNA damage checkpoint and thereby to apoptosis or cell cycle arrest, whereas a second barrier is mediated by oncogene-induced senescence. The relationship between these two barriers, if any, has not been elucidated. Here we show that oncogene-induced senescence is associated with signs of DNA replication stress, including prematurely terminated DNA replication forks and DNA double-strand breaks. Inhibiting the DNA double-strand break response kinase ataxia telangiectasia mutated (ATM) suppressed the induction of senescence and in a mouse model led to increased tumour size and invasiveness. Analysis of human precancerous lesions further indicated that DNA damage and senescence markers cosegregate closely. Thus, senescence in human preneoplastic lesions is a manifestation of oncogene-induced DNA replication stress and, together with apoptosis, provides a barrier to malignant progression.


Assuntos
Transformação Celular Neoplásica/genética , Senescência Celular/genética , Dano ao DNA , Oncogenes , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular , Ciclina E/genética , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , DNA , Replicação do DNA , Genes mos , Humanos , Camundongos , Invasividade Neoplásica/genética , Proteínas Nucleares/genética , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia
4.
Nat Cell Biol ; 4(12): 998-1002, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12447382

RESUMO

53BP1 is a conserved nuclear protein that is implicated in the DNA damage response. After irradiation, 53BP1 localizes rapidly to nuclear foci, which represent sites of DNA double strand breaks, but its precise function is unclear. Using small interference RNA (siRNA), we demonstrate that 53BP1 functions as a DNA damage checkpoint protein. 53BP1 is required for at least a subset of ataxia telangiectasia-mutated (ATM)-dependent phosphorylation events at sites of DNA breaks and for cell cycle arrest at the G2-M interphase after exposure to irradiation. Interestingly, in cancer cell lines expressing mutant p53, 53BP1 was localized to distinct nuclear foci and ATM-dependent phosphorylation of Chk2 at Thr 68 was detected, even in the absence of irradiation. In addition, Chk2 was phosphorylated at Thr 68 in more than 50% of surgically resected lung and breast tumour specimens from otherwise untreated patients [corrected]. We conclude that the constitutive activation of the DNA damage checkpoint pathway may be linked to the high frequency of p53 mutations in human cancer, as p53 is a downstream target of Chk2 and ATM.


Assuntos
Proteínas de Transporte/genética , Ciclo Celular/genética , Dano ao DNA/genética , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular , Fosfoproteínas , Proteínas Serina-Treonina Quinases/genética , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Genes p53 , Humanos , Mutação , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
5.
Nature ; 434(7035): 864-70, 2005 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-15829956

RESUMO

During the evolution of cancer, the incipient tumour experiences 'oncogenic stress', which evokes a counter-response to eliminate such hazardous cells. However, the nature of this stress remains elusive, as does the inducible anti-cancer barrier that elicits growth arrest or cell death. Here we show that in clinical specimens from different stages of human tumours of the urinary bladder, breast, lung and colon, the early precursor lesions (but not normal tissues) commonly express markers of an activated DNA damage response. These include phosphorylated kinases ATM and Chk2, and phosphorylated histone H2AX and p53. Similar checkpoint responses were induced in cultured cells upon expression of different oncogenes that deregulate DNA replication. Together with genetic analyses, including a genome-wide assessment of allelic imbalances, our data indicate that early in tumorigenesis (before genomic instability and malignant conversion), human cells activate an ATR/ATM-regulated DNA damage response network that delays or prevents cancer. Mutations compromising this checkpoint, including defects in the ATM-Chk2-p53 pathway, might allow cell proliferation, survival, increased genomic instability and tumour progression.


Assuntos
Transformação Celular Neoplásica , Dano ao DNA , Neoplasias/patologia , Neoplasias/prevenção & controle , Desequilíbrio Alélico/genética , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Quinase do Ponto de Checagem 2 , Ciclina E/genética , Ciclina E/metabolismo , Dano ao DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição E2F , Ativação Enzimática , Genes p53/genética , Instabilidade Genômica , Humanos , Mutação/genética , Neoplasias/enzimologia , Neoplasias/genética , Oncogenes/genética , Oncogenes/fisiologia , Fosforilação , Polimorfismo de Nucleotídeo Único/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Neoplasias da Bexiga Urinária/enzimologia , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo
6.
Electrophoresis ; 31(16): 2714-21, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20717991

RESUMO

The anticancer drug belinostat is a hydroxamate histone deacetylase inhibitor that has shown significant antitumour activity in various tumour models and also in clinical trials. In this study, we utilized a proteomic approach in order to evaluate the effect of this drug on protein expression in the human colon cancer cell line HCT116. Protein extracts from untreated HCT116 cells, and cells grown for 24 h in the presence of 1 and 10 muM belinostat were analysed by 2-D gel electrophoresis. Proteins were visualized by colloidal Coomassie blue staining and quantitative analysis of gel images revealed 45 unique differentially expressed proteins that were identified by LC-MSMS analysis. Among these proteins, of particular interest are the downregulated proteins nucleophosmin and stratifin, and the upregulated proteins nucleolin, gelsolin, heterogeneous nuclear ribonucleoprotein K, annexin 1, and HSP90B that all were related to the proto-oncogene proteins p53, Myc, activator protein 1, and c-fos protein. The modulation of these proteins is consistent with the observations that belinostat is able to inhibit clonogenic cell growth of HCT116 cells and the biological role of these proteins will be discussed.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Perfilação da Expressão Gênica , Inibidores de Histona Desacetilases/uso terapêutico , Ácidos Hidroxâmicos/uso terapêutico , Proteoma/química , Western Blotting , Morte Celular , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Ensaio de Unidades Formadoras de Colônias/métodos , Biologia Computacional , Eletroforese em Gel Bidimensional/métodos , Histona Desacetilases/isolamento & purificação , Histona Desacetilases/metabolismo , Humanos , Proto-Oncogene Mas , Proto-Oncogenes/efeitos dos fármacos , Sulfonamidas
7.
BMC Cancer ; 10: 677, 2010 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-21144000

RESUMO

BACKGROUND: Inhibitors of nicotinamide phosphoribosyltransferase (NAMPT) are promising cancer drugs currently in clinical trials in oncology, including APO866, CHS-828 and the CHS-828 prodrug EB1627/GMX1777, but cancer cell resistance to these drugs has not been studied in detail. METHODS: Here, we introduce an analogue of CHS-828 called TP201565 with increased potency in cellular assays. Further, we describe and characterize a panel of cell lines with acquired stable resistance towards several NAMPT inhibitors of 18 to 20,000 fold compared to their parental cell lines. RESULTS: We find that 4 out of 5 of the resistant sublines display mutations of NAMPT located in the vicinity of the active site or in the dimer interface of NAMPT. Furthermore, we show that these mutations are responsible for the resistance observed. All the resistant cell lines formed xenograft tumours in vivo. Also, we confirm CHS-828 and TP201565 as competitive inhibitors of NAMPT through docking studies and by NAMPT precipitation from cellular lysate by an analogue of TP201565 linked to sepharose. The NAMPT precipitation could be inhibited by addition of APO866. CONCLUSION: We found that CHS-828 and TP201565 are competitive inhibitors of NAMPT and that acquired resistance towards NAMPT inhibitors can be expected primarily to be caused by mutations in NAMPT.


Assuntos
Acrilamidas/farmacologia , Antineoplásicos/farmacologia , Cianetos/farmacologia , Citocinas/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Guanidinas/farmacologia , Mutação , Neoplasias/tratamento farmacológico , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Piperidinas/farmacologia , Acrilamidas/metabolismo , Animais , Antineoplásicos/metabolismo , Sítios de Ligação , Ligação Competitiva , Domínio Catalítico , Linhagem Celular Tumoral , Cianetos/metabolismo , Citocinas/química , Citocinas/genética , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/metabolismo , Feminino , Guanidinas/metabolismo , Células HCT116 , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos Nus , Modelos Moleculares , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Nicotinamida Fosforribosiltransferase/química , Nicotinamida Fosforribosiltransferase/genética , Nicotinamida Fosforribosiltransferase/metabolismo , Piperidinas/metabolismo , Conformação Proteica , Fatores de Tempo , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Histopathology ; 54(6): 688-98, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19438744

RESUMO

AIMS: Histone deacetylase (HDAC) inhibitors are novel therapeutics in the treatment of peripheral T-cell lymphoma, unspecified (PTCL) and diffuse large B-cell lymphoma (DLBCL), where, for unknown reasons, T-cell malignancies appear to be more sensitive than B-cell malignancies. The aim was to determine HDAC expression in DLBCL and PTCL which has not previously been investigated. METHODS AND RESULTS: The expression of HDAC1, HDAC2, HDAC6 and acetylated histone H4 was examined immunohistochemically in 31 DLBCL and 45 PTCL. All four markers showed high expression in both DLBCL and PTCL compared with normal lymphoid tissue. HDAC1 was more abundantly expressed in PTCL than in DLBCL (P = 0.0046), whereas acetylated H4 was more frequent in DLBCL (P < 0.0001), the latter suggesting a mechanism for T-cell lymphoma sensitivity to HDAC inhibitors. Moderate to strong HDAC6 expression was significantly correlated with favourable outcome (P = 0.016) in DLBCL patients, whereas the opposite effect was observed in PTCL patients (P < 0.0001). The other markers did not correlate with survival (P > 0.05). CONCLUSIONS: HDAC1, HDAC2, HDAC6 and acetylated H4 are overexpressed in DLBCL and PTCL relative to normal lymphoid tissue. Furthermore, HDAC6 may be an important prognostic marker associated with favourable outcome in DLBCL and a more aggressive course in PTCL.


Assuntos
Histona Desacetilases/metabolismo , Histonas/metabolismo , Linfoma Difuso de Grandes Células B/enzimologia , Linfoma de Células T Periférico/enzimologia , Proteínas Repressoras/metabolismo , Acetilação , Linhagem Celular Tumoral , Histona Desacetilase 1 , Histona Desacetilase 2 , Desacetilase 6 de Histona , Humanos , Imuno-Histoquímica , Linfoma Difuso de Grandes Células B/metabolismo , Linfoma Difuso de Grandes Células B/patologia , Linfoma de Células T Periférico/metabolismo , Linfoma de Células T Periférico/patologia
9.
Anticancer Drugs ; 20(8): 682-92, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19606018

RESUMO

Belinostat is a hydroxamate-type histone deactylase inhibitor (HDACi), which has recently entered phase I and II clinical trials. Microarray-based analysis of belinostat-treated cell lines showed an impact on genes associated with the G2/M phase of the cell cycle and downregulation of the aurora kinase pathway. Expression of 25 dysregulated genes was measured in eight differentially sensitive cell lines using a novel high-throughput assay that combines multiplex reverse transcriptase-PCR and fluorescence capillary electrophoresis. Sensitivity to belinostat and the magnitude of changes in overall gene modulation were significantly correlated. A belinostat-gene profile was specific for HDACi in three cell lines when compared with equipotent concentrations of four mechanistically different chemotherapeutic agents: 5-fluorouracil, cisplatin, paclitaxel, and thiotepa. Belinostat- and trichostatin A (HDACi)-induced gene responses were highly correlated with each other, but not with the limited changes in response to the other non-HDACi agents. Moreover, belinostat treatment of mice bearing human xenografts showed that the preponderance of selected genes were also modulated in vivo, more extensively in a drug-sensitive tumor than a more resistant model. We have demonstrated a gene signature that is selectively regulated by HDACi when compared with other clinical agents allowing us to distinguish HDACi responses from those related to other mechanisms.


Assuntos
Inibidores Enzimáticos/farmacologia , Perfilação da Expressão Gênica , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Antineoplásicos/farmacologia , Aurora Quinases , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas Cromossômicas não Histona/genética , Regulação para Baixo/genética , Inibidores Enzimáticos/uso terapêutico , Feminino , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Células HCT116 , Humanos , Ácidos Hidroxâmicos/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Sulfonamidas , Regulação para Cima/genética , Proteína ran de Ligação ao GTP/genética
10.
Toxicology ; 255(1-2): 72-9, 2009 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-19010377

RESUMO

Anthracycline-induced cardiomyopathy is a major problem in anti-cancer therapy. The only approved agent for alleviating this serious dose limiting side effect is ICRF-187 (dexrazoxane). The current thinking is that the ring-opened hydrolysis product of this agent, ADR-925, which is formed inside cardiomyocytes, removes iron from its complexes with anthracyclines, hereby reducing the concentration of highly toxic iron-anthracycline complexes that damage cardiomyocytes by semiquinone redox recycling and the production of free radicals. However, the 2 carbon linker ICRF-187 is also is a catalytic inhibitor of topoisomerase II, resulting in the risk of additional myelosuppression in patients receiving ICRF-187 as a cardioprotectant in combination with doxorubicin. The development of a topoisomerase II-inactive iron chelating compound thus appeared attractive. In the present paper we evaluate the topoisomerase II-inactive 3 carbon linker bisdioxopiperazine analog ICRF-161 as a cardioprotectant. We demonstrate that this compound does chelate iron and protects against doxorubicin-induced LDH release from primary rat cardiomyocytes in vitro, similarly to ICRF-187. The compound does not target topoisomerase II in vitro or in cells, it is well tolerated and shows similar exposure to ICRF-187 in rodents, and it does not induce myelosuppression when given at high doses to mice as opposed to ICRF-187. However, when tested in a model of chronic anthracycline-induced cardiomyopathy in spontaneously hypertensive rats, ICRF-161 was not capable of protecting against the cardiotoxic effects of doxorubicin. Modulation of the activity of the beta isoform of the topoisomerase II enzyme by ICRF-187 has recently been proposed as the mechanism behind its cardioprotection. This concept is thus supported by the present study in that iron chelation alone does not appear to be sufficient for protection against anthracycline-induced cardiomyopathy.


Assuntos
Antibióticos Antineoplásicos/antagonistas & inibidores , Antibióticos Antineoplásicos/toxicidade , Antineoplásicos/farmacologia , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/prevenção & controle , DNA Topoisomerases Tipo II/metabolismo , Doxorrubicina/antagonistas & inibidores , Doxorrubicina/toxicidade , Razoxano/farmacologia , Animais , Animais Recém-Nascidos , Antineoplásicos/farmacocinética , Cardiomiopatias/patologia , Ensaio de Unidades Formadoras de Colônias , Crithidia fasciculata/metabolismo , DNA/efeitos dos fármacos , Compostos Férricos/farmacologia , Estimativa de Kaplan-Meier , L-Lactato Desidrogenase/metabolismo , Camundongos , Mitocôndrias Cardíacas/efeitos dos fármacos , Miocárdio/patologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Ratos , Ratos Endogâmicos SHR , Razoxano/farmacocinética , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/patologia , Troponina I/metabolismo
11.
Biochem J ; 409(2): 581-9, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17868033

RESUMO

The human HDAC (histone deacetylase) family, a well-validated anticancer target, plays a key role in the control of gene expression through regulation of transcription. While HDACs can be subdivided into three main classes, the class I, class II and class III HDACs (sirtuins), it is presently unclear whether inhibiting multiple HDACs using pan-HDAC inhibitors, or targeting specific isoforms that show aberrant levels in tumours, will prove more effective as an anticancer strategy in the clinic. To address the above issues, we have tested a number of clinically relevant HDACis (HDAC inhibitors) against a panel of rhHDAC (recombinant human HDAC) isoforms. Eight rhHDACs were expressed using a baculoviral system, and a Fluor de Lystrade mark (Biomol International) HDAC assay was optimized for each purified isoform. The potency and selectivity of ten HDACs on class I isoforms (rhHDAC1, rhHDAC2, rhHDAC3 and rhHDAC8) and class II HDAC isoforms (rhHDAC4, rhHDAC6, rhHDAC7 and rhHDAC9) was determined. MS-275 was HDAC1-selective, MGCD0103 was HDAC1- and HDAC2-selective, apicidin was HDAC2- and HDAC3-selective and valproic acid was a specific inhibitor of class I HDACs. The hydroxamic acid-derived compounds (trichostatin A, NVP-LAQ824, panobinostat, ITF2357, vorinostat and belinostat) were potent pan-HDAC inhibitors. The growth-inhibitory effect of the HDACis on HeLa cells showed that both pan-HDAC and class-I-specific inhibitors inhibited cell growth. The results also showed that both pan-HDAC and class-I-specific inhibitor treatment resulted in increased acetylation of histones, but only pan-HDAC inhibitor treatment resulted in increased tubulin acetylation, which is in agreement with their activity towards the HDAC6 isoform.


Assuntos
Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Acetilação , Proliferação de Células , Clonagem Molecular , Inibidores Enzimáticos/metabolismo , Células HeLa , Histona Desacetilases/classificação , Histona Desacetilases/metabolismo , Humanos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
Tumori ; 95(3): 273-82, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19688963

RESUMO

An accidental extravasation of anthracycline-containing chemotherapy is a feared complication that may lead to necrosis and severe tissue destruction. For four decades, much effort has been done to prevent and treat this devastating condition. Savene has recently been proved to be very effective, and is the only approved treatment against anthracyline extravasation. It is thus now widely recommended. The present article represents a comprehensive review of, and historical insight to, the experimental and clinical studies of surgical and non-surgical treatments of extravasation during forty years of clinical anthracycline treatment.


Assuntos
Antraciclinas/efeitos adversos , Antibióticos Antineoplásicos/efeitos adversos , Extravasamento de Materiais Terapêuticos e Diagnósticos/terapia , Dermatopatias/induzido quimicamente , Dermatopatias/terapia , Corticosteroides/uso terapêutico , Animais , Antraciclinas/administração & dosagem , Antibióticos Antineoplásicos/administração & dosagem , Bicarbonatos/uso terapêutico , Dimetil Sulfóxido/uso terapêutico , Extravasamento de Materiais Terapêuticos e Diagnósticos/tratamento farmacológico , Extravasamento de Materiais Terapêuticos e Diagnósticos/etiologia , Extravasamento de Materiais Terapêuticos e Diagnósticos/cirurgia , Fator 2 de Crescimento de Fibroblastos/uso terapêutico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Humanos , Hialuronoglucosaminidase/uso terapêutico , Necrose/induzido quimicamente , Necrose/terapia , Razoxano/uso terapêutico , Dermatopatias/tratamento farmacológico , Dermatopatias/cirurgia , Úlcera/induzido quimicamente , Úlcera/terapia , alfa-Tocoferol/uso terapêutico
13.
Mol Cancer ; 7: 70, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18789133

RESUMO

BACKGROUND: Histone acetylation is an epigenetic modification involved in the regulation of gene expression, balanced by histone acetyl transferases and histone deacetylase (HDAC) enzymes. HDAC inhibitors (HDACi) induce growth arrest and cell death in transformed cells, and are currently in many clinical cancer trials. The transcriptional response to HDACi is complex, as is the response to HDAC isoform knockdown (KD). Here, we describe for the first time in a human cancer cell line, a transcriptional comparison of treatment by two structurally unrelated HDACi; belinostat and valproic acid with the KD of HDAC1, 2 and 3 isoforms. RESULTS: HDAC KD showed anti-proliferative effects, although to a lesser extent than HDACi treatment. Moreover, we found a 2-fold increased resistance of HDAC1 knockdown cells to belinostat, suggesting this isoenzyme as a selective target. While both HDACi treatment and individual class I HDAC KD produce significant transcriptional effects, three-times higher for HDACi, the gene-expression profiles of class I HDAC KD compared with that obtained by HDACi treatment exhibited less than 4% of altered genes in common between the two modes of inhibition. Further, cell-specific effects of HDAC KD are evident by comparison with a recent study in a different cell line. CONCLUSION: The increased resistance to belinostat in response to HDAC1 depletion indicates the possibility of using this isoform as a predictive biomarker of response to HDACi treatment. Further, the transcriptional response to chemical inhibition of HDACs is very different from that of KD of individual class I HDAC isoforms. These data suggest that the anti-tumor effect of HDACi is indeed linked to class I inhibition, but may be more complex than simply targeting individual HDAC enzymes.


Assuntos
Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos/farmacologia , Ácido Valproico/farmacologia , Sobrevivência Celular , Regulação Enzimológica da Expressão Gênica , Células HeLa , Histona Desacetilase 1 , Histona Desacetilase 2 , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Sulfonamidas , Transcrição Gênica
14.
Biochem Biophys Res Commun ; 367(4): 799-804, 2008 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-18201551

RESUMO

Malignant cells display increased demands for energy production and DNA repair. Nicotinamide adenine dinucleotide (NAD) is required for both processes and is also continuously degraded by cellular enzymes. Nicotinamide phosphoribosyltransferase (Nampt) is a crucial factor in the resynthesis of NAD, and thus in cancer cell survival. Here, we establish the cytotoxic mechanism of action of the small molecule inhibitor CHS-828 to result from impaired synthesis of NAD. Initially, we detected cross-resistance in cells between CHS-828 and a known inhibitor of Nampt, FK866, a compound of a structurally different class. We then showed that nicotinamide protects against CHS-828-mediated cytotoxicity. Finally, we observed that treatment with CHS-828 depletes cellular NAD levels in sensitive cancer cells. In conclusion, these results strongly suggest that, like FK866, CHS-828 kills cancer cells by depleting NAD.


Assuntos
Acrilamidas/administração & dosagem , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Cianetos/administração & dosagem , Guanidinas/administração & dosagem , Neoplasias Pulmonares/metabolismo , NAD/metabolismo , Piperidinas/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Neoplasias Pulmonares/tratamento farmacológico
15.
APMIS ; 116(5): 382-92, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18452428

RESUMO

Histone deacetylase (HDAC) inhibition is a novel entity in medical oncology, and several HDAC inhibitors are in clinical trials. One of them is the hydroxamic acid belinostat (PXD101) that has demonstrated therapeutic efficacy for several clinical indications. Acetylation of histones is a key event after treatment with HDAC inhibitors, and could thus be used as a marker for monitoring cellular response to HDAC inhibitor treatment. Here we describe the utility of a newly described monoclonal antibody against acetylated H4 for immunohistochemistry on paraffin-embedded fine needle biopsies from nude mice carrying A2780 human ovarian cancer xenografts. Acetylated H4 was monitored in vivo by immunohistochemistry during treatment with belinostat, and compared with pharmacokinetics in plasma and tumor tissue. We found an increased level of acetylated H4 15 min after a single treatment (200 mg/kg i.v.) with maximum level reached after 1 h. H4 acetylation intensity reflected the belinostat concentration in plasma and tumor tissue. The threshold level for belinostat activity, indicated by acetylated H4, correlated with belinostat plasma concentrations above 1,000 ng/ml. In conclusion, examination of H4 acetylation in fine needle biopsies using the T25 antibody may prove useful in monitoring HDAC inhibitor efficacy in clinical trials involving humans with solid tumors.


Assuntos
Antineoplásicos/uso terapêutico , Histonas/metabolismo , Ácidos Hidroxâmicos/uso terapêutico , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Acetilação/efeitos dos fármacos , Animais , Anticorpos Monoclonais , Antineoplásicos/sangue , Antineoplásicos/farmacocinética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Linhagem Celular Tumoral , Feminino , Células HCT116 , Inibidores de Histona Desacetilases , Histona Desacetilases/imunologia , Histonas/imunologia , Humanos , Ácidos Hidroxâmicos/sangue , Ácidos Hidroxâmicos/farmacocinética , Leucemia P388/tratamento farmacológico , Leucemia P388/enzimologia , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Neoplasias Hepáticas Experimentais/enzimologia , Masculino , Camundongos , Camundongos Nus , Neoplasias Experimentais/enzimologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/enzimologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/enzimologia , Sulfonamidas , Transplante Heterólogo
16.
Exp Dermatol ; 17(11): 972-4, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18616577

RESUMO

Retinoids and the histone deacetylase inhibitor valproic acid have shown anticancer properties, but the photocarcinogenic or photoprotective effect is unclear. Therefore, we investigated whether a topical formulation of valproic acid is photocarcinogenic or photoprotective in hairless female C3.Cg/TifBomTac immunocompetent mice exposed to simulated solar radiation (SSR) and whether valproic acid changes the effect of the retinoids: tazarotene and isotretinoin. The products were applied on the dorsal skin of 400 mice (five times weekly) followed by SSR (three times weekly) 3-4 h after the application. This was performed during 12 months or until death. Tumors appeared sooner in groups treated with tazarotene and isotretinoin compared with that of the group treated with valproic acid and the control group. The present study shows that valproic acid alone is not photocarcinogenic or photoprotective in hairless mice. When valproic acid is combined with tazarotene or isotretinoin, it does not change their photocarcinogenicity significantly.


Assuntos
Isotretinoína/toxicidade , Ácidos Nicotínicos/toxicidade , Neoplasias Cutâneas/patologia , Ácido Valproico/toxicidade , Administração Cutânea , Animais , Carcinógenos/administração & dosagem , Carcinógenos/toxicidade , Fármacos Dermatológicos/administração & dosagem , Fármacos Dermatológicos/toxicidade , Relação Dose-Resposta à Radiação , Feminino , Isotretinoína/administração & dosagem , Estimativa de Kaplan-Meier , Camundongos , Camundongos Pelados , Ácidos Nicotínicos/administração & dosagem , Processos Fotoquímicos , Pele/efeitos dos fármacos , Pele/patologia , Pele/efeitos da radiação , Neoplasias Cutâneas/induzido quimicamente , Raios Ultravioleta , Ácido Valproico/administração & dosagem
17.
Cancer Chemother Pharmacol ; 62(3): 433-7, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17960383

RESUMO

PURPOSE: Histone deacetylases (HDAC) are involved in the regulation of gene transcription. Aberrant HDAC activity has been associated with tumorigenesis, and, therefore, HDACs are potential targets for the treatment of cancers, including tumors of the central nervous system (CNS). Belinostat is a novel, potent, pan-HDAC inhibitor with antiproliferative activity on a wide variety of tumor cell lines. We studied the cerebrospinal fluid (CSF) penetration of intravenous (IV) belinostat in a non-human primate model as a surrogate for blood:brain barrier penetration. DESIGN: Five adult rhesus monkeys received increasing doses of belinostat (10-60 mg/kg) as a 30-min IV infusion. Serial blood and CSF samples were collected over 48 h. Plasma and CSF concentrations of belinostat were quantified with an LC/MS/MS assay. Pharmacokinetic parameters were calculated using non-compartmental methods, and CSF penetration is expressed as the ratio of the area under the concentration-time curve (AUC) in CSF to the AUC in plasma. RESULTS: Belinostat was cleared rapidly from plasma with a half-life of 1.0 h, a mean residence time of 0.47 h, and a clearance of 425 ml/min/m(2). CSF penetration of belinostat was limited. CSF drug exposure was <1% of plasma drug exposure and <10% of free (non-protein bound) plasma drug exposure. CONCLUSION: IV belinostat is rapidly cleared from plasma and has limited penetration into the CSF.


Assuntos
Inibidores Enzimáticos , Inibidores de Histona Desacetilases , Ácidos Hidroxâmicos , Animais , Área Sob a Curva , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/líquido cefalorraquidiano , Ácidos Hidroxâmicos/administração & dosagem , Ácidos Hidroxâmicos/sangue , Ácidos Hidroxâmicos/líquido cefalorraquidiano , Infusões Intravenosas , Macaca mulatta , Masculino , Taxa de Depuração Metabólica , Sulfonamidas
18.
Mol Cell Biol ; 25(9): 3553-62, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15831461

RESUMO

Human checkpoint kinase 1 (Chk1) is an essential kinase required to preserve genome stability. Here, we show that Chk1 inhibition by two distinct drugs, UCN-01 and CEP-3891, or by Chk1 small interfering RNA (siRNA) leads to phosphorylation of ATR targets. Chk1-inhibition triggered rapid, pan-nuclear phosphorylation of histone H2AX, p53, Smc1, replication protein A, and Chk1 itself in human S-phase cells. These phosphorylations were inhibited by ATR siRNA and caffeine, but they occurred independently of ATM. Chk1 inhibition also caused an increased initiation of DNA replication, which was accompanied by increased amounts of nonextractable RPA protein, formation of single-stranded DNA, and induction of DNA strand breaks. Moreover, these responses were prevented by siRNA-mediated downregulation of Cdk2 or the replication initiation protein Cdc45, or by addition of the CDK inhibitor roscovitine. We propose that Chk1 is required during normal S phase to avoid aberrantly increased initiation of DNA replication, thereby protecting against DNA breakage. These results may help explain why Chk1 is an essential kinase and should be taken into account when drugs to inhibit this kinase are considered for use in cancer treatment.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Dano ao DNA/fisiologia , Replicação do DNA/fisiologia , Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/fisiologia , Estaurosporina/análogos & derivados , Proteínas Mutadas de Ataxia Telangiectasia , Cafeína/farmacologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Proteínas Cromossômicas não Histona/metabolismo , Replicação do DNA/efeitos dos fármacos , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Humanos , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/genética , Proteínas Quinases/fisiologia , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Purinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Proteína de Replicação A , Roscovitina , Estaurosporina/farmacologia , Proteína Supressora de Tumor p53/metabolismo
19.
Cancer Res ; 66(21): 10258-63, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17079443

RESUMO

Loss of G(1)-S control and aberrations of the p16(Ink4a)-cyclin D1/cyclin-dependent kinase (CDK) 4(6)-pRb-E2F-cyclin E/CDK2 pathway are common in human cancer. Previous studies showed that oncogene-induced aberrant proliferation, such as on cyclin E overexpression, causes DNA damage and checkpoint activation. Here, we show that, in a series of human colorectal adenomas, those with deregulation of cyclin D1 and/or p16(Ink4a) showed little evidence of constitutive DNA damage response (DDR), contrary to cyclin E-overexpressing higher-grade cases. These observations were consistent with diverse cell culture models with differential defects of retinoblastoma pathway components, as overexpression of cyclin D1 or lack of p16(Ink4a), either alone or combined, did not elicit detectable DDR. In contrast, inactivation of pRb, the key component of the pathway, activated the DDR in cultured human or mouse cells, analogous to elevated cyclin E. These results highlight differential effect of diverse oncogenic events on driving the 'cancer cell cycles' and their ability to deregulate the replication-driving CDK2 kinase and to alarm the DDR as a potential anticancer barrier in accordance with their hierarchical positions along the retinoblastoma pathway. Our data provide new insights into oncogene-evoked DDR in human tumorigenesis, with potential implications for individualized management of tumors with elevated cyclin D1 versus cyclin E, due to their distinct clinical variables and biological behavior.


Assuntos
Dano ao DNA , Neoplasias/genética , Proteína do Retinoblastoma/fisiologia , Linhagem Celular Tumoral , Ciclina D1/fisiologia , Ciclina E/fisiologia , Quinase 2 Dependente de Ciclina/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Fator de Transcrição E2F1/fisiologia , Genes p53 , Humanos , Neoplasias/etiologia
20.
Mol Cancer Res ; 4(5): 293-302, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16687484

RESUMO

Matrix metalloproteinase-9 (MMP-9) is up-regulated in macrophages in various human cancer types. In human colon cancer, MMP-9 is expressed in a macrophage subpopulation located at the tumor edge, indicating a specific induction of MMP-9 in macrophages in direct association with cancer invasion. To test whether MMP-9 is also induced in tumor edge macrophages in metastases from colorectal adenocarcinomas, we have compared the expression pattern of MMP-9 in primary colorectal adenocarcinomas (n = 15) with that in liver metastases (n = 15) and local lymph node metastases (n = 7) from the same patients by in situ hybridization and immunohistochemistry. In all the colorectal adenocarcinomas, the expression of MMP-9 mRNA and immunoreactivity in macrophages was located at the invasive front. In contrast, only 3 of the 15 liver metastases had MMP-9 mRNA and immunoreactivity at the periphery, and this expression was confined to small foci of macrophages located either among lymphocytes or in a dense desmoplastic stroma. Expression of MMP-9 mRNA and immunoreactivity was in all liver metastases seen in macrophages located in the lumen of malignant glandular structures and in central necrotic tissue. In all the 7 lymph node metastases, MMP-9 mRNA and immunoreactivity was seen in macrophages located in the stromal tissue surrounding the metastases. We conclude that MMP-9 is not up-regulated in tumor edge macrophages in liver metastases like in their primary tumor and local lymph node metastases, suggesting that disseminating colorectal cancer cells can adopt alternative proteolytic mechanisms for invasion depending on the local microenvironment.


Assuntos
Adenocarcinoma/enzimologia , Neoplasias Colorretais/enzimologia , Metaloproteinase 9 da Matriz/biossíntese , Adenocarcinoma/genética , Adenocarcinoma/patologia , Idoso , Idoso de 80 Anos ou mais , Anticorpos/química , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Feminino , Humanos , Técnicas Imunoenzimáticas/métodos , Hibridização In Situ/métodos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/secundário , Metástase Linfática , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica , Metástase Neoplásica , RNA Mensageiro/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA