Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Ann Oncol ; 31(8): 1040-1045, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32387456

RESUMO

BACKGROUND: Cell entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) depends on binding of the viral spike (S) proteins to angiotensin-converting enzyme 2 and on S protein priming by TMPRSS2. Inhibition of TMPRSS2 may work to block or decrease the severity of SARS-CoV-2 infections. Intriguingly, TMPRSS2 is an androgen-regulated gene that is up-regulated in prostate cancer where it supports tumor progression and is involved in a frequent genetic translocation with the ERG gene. First- or second-generation androgen-deprivation therapies (ADTs) decrease the levels of TMPRSS2. Here we put forward the hypothesis that ADTs may protect patients affected by prostate cancer from SARS-CoV-2 infections. MATERIALS AND METHODS: We extracted data regarding 9280 patients (4532 males) with laboratory-confirmed SARS-CoV-2 infection from 68 hospitals in Veneto, one of the Italian regions that was most affected by the coronavirus disease 2019 (COVID-19) pandemic. The parameters used for each COVID-19-positive patient were sex, hospitalization, admission to intensive care unit, death, tumor diagnosis, prostate cancer diagnosis, and ADT. RESULTS: There were evaluable 9280 SARS-CoV-2-positive patients in Veneto on 1 April 2020. Overall, males developed more severe complications, were more frequently hospitalized, and had a worse clinical outcome than females. Considering only the Veneto male population (2.4 million men), 0.2% and 0.3% of non-cancer and cancer patients, respectively, tested positive for SARS-CoV-2. Comparing the total number of SARS-CoV-2-positive cases, prostate cancer patients receiving ADT had a significantly lower risk of SARS-CoV-2 infection compared with patients who did not receive ADT (OR 4.05; 95% CI 1.55-10.59). A greater difference was found comparing prostate cancer patients receiving ADT with patients with any other type of cancer (OR 4.86; 95% CI 1.88-12.56). CONCLUSION: Our data suggest that cancer patients have an increased risk of SARS-CoV-2 infections compared with non-cancer patients. However, prostate cancer patients receiving ADT appear to be partially protected from SARS-CoV-2 infections.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Betacoronavirus , Infecções por Coronavirus/prevenção & controle , Pandemias/prevenção & controle , Pneumonia Viral/prevenção & controle , Vigilância da População , Neoplasias da Próstata/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , COVID-19 , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/epidemiologia , Humanos , Itália/epidemiologia , Masculino , Pessoa de Meia-Idade , Pneumonia Viral/diagnóstico , Pneumonia Viral/epidemiologia , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/epidemiologia , Fatores de Risco , SARS-CoV-2
2.
Invest New Drugs ; 35(6): 766-772, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28317088

RESUMO

Purpose To establish a recommended phase II dose (RP2D) for the oral smoothened inhibitor sonidegib in combination with paclitaxel; secondary objectives include evaluation of safety, tolerability, markers of Hedgehog (Hh) signaling and preliminary antitumor activity. Methods Patients with advanced solid tumors were enrolled in cohorts of escalating sonidegib dose levels (400mg, 600mg and 800mg orally, once daily on days 1-28) in combination with paclitaxel 80 mg/m2 on days 1, 8 and 15 in 4-weekly cycles. Dose-limiting toxicities (DLTs) were assessed using CTCAE v4. Once the RP2D was defined, patients with advanced ovarian carcinoma were treated at this dose level in an expansion phase. Biomarkers of Hh signaling were assessed by immunohistochemistry in archival tissue and antitumor activity evaluated using RECIST 1.1. Results 18 patients were treated: 3 at 400 mg, 3 at 600 mg and 12 at 800 mg sonidegib. Only one patient treated at 800 mg presented a DLT (prolonged neutropenia resulting in failure to receive 75% of the planned sonidegib dose). However, 4 of 12 patients treated at 800 mg had their sonidegib dose reduced for toxicity after cycle 1. Hh biomarker (SHH, Patched, SMO and GLI1) staining did not correlate with clinical activity. Best response was partial response in 3 patients (2 ovarian, 1 breast cancer) and stable disease >4 cycles in 3 patients (2 ovarian, 1 anal cancer). Conclusions The combination of sonidegib and paclitaxel is tolerable and evidence of antitumor activity was identified. The RP2D of sonidegib was 800 mg in combination with paclitaxel 80mg/m2.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Receptor Smoothened/antagonistas & inibidores , Administração Oral , Idoso , Biomarcadores Tumorais , Compostos de Bifenilo/administração & dosagem , Relação Dose-Resposta a Droga , Feminino , Seguimentos , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Neoplasias/metabolismo , Neoplasias/patologia , Paclitaxel/administração & dosagem , Prognóstico , Piridinas/administração & dosagem
3.
Nucleic Acids Res ; 30(12): 2701-9, 2002 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12060688

RESUMO

Altered expression of c-myc is implicated in pathogenesis and progression of many human cancers. Triple helix-forming oligonucleotides (TFOs) directed to a polypurine/polypyrimidine sequence in a critical regulatory region near the c-myc P2 promoter have been shown to inhibit c-myc transcription in vitro and in cells. However, these guanine-rich TFOs had moderate binding affinity and required high concentrations for activity. The 23 bp myc P2 sequence is split equally into AT- and GC-rich tracts. Gel mobility analysis of a series of short TFOs directed in parallel and anti-parallel orientation to the purine strand of each tract showed that only parallel CT and anti-parallel GT TFOs formed stable triplex on the AT- and GC-rich tracts, respectively. A novel full-length GTC TFO was designed to bind simultaneously in parallel and anti-parallel orientation to the polypurine strand. Gel-shift and footprinting assays showed that the new TFO formed a triple helix in physiological conditions with significantly higher affinity than an anti-parallel TFO. Protein-binding assays showed that 1 microM GTC TFO inhibited binding of nuclear transcription factors to the P2 promoter sequence. The novel TFO can be developed into a potent antigene agent, and its design strategy applied to similar genomic sequences, thus expanding the TFO repertoire.


Assuntos
DNA/síntese química , DNA/farmacologia , Genes myc , Oligodesoxirribonucleotídeos/farmacologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/genética , Sequência Rica em At , Sequência de Bases , Citosina/química , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Exodesoxirribonucleases/química , Sequência Rica em GC , Guanina/química , Humanos , Oligodesoxirribonucleotídeos/síntese química , Oligodesoxirribonucleotídeos/metabolismo , Timidina/química , Células Tumorais Cultivadas
4.
Cancer Res ; 51(7): 1829-35, 1991 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-1706219

RESUMO

The effects of fludarabine triphosphate (Fara-ATP), 1-beta-D-arabinofuranosylcytosine 5'-triphosphate (ara-CTP), and aphidicolin on primer RNA and DNA synthesis in human CCRF-CEM leukemia cells were investigated. RNA-primed Okazaki fragment synthesis was monitored by first incubating whole cell lysates for 10 min in the presence or absence of the compound and then following the incorporation of [alpha-32P]ATP and [3H]dTTP into the primer RNA and DNA portions, respectively, of the Okazaki fragments. In whole cell lysates the degree of DNA synthesis inhibition induced by Fara-ATP was directly related to the extent of primer RNA synthesis inhibition over the entire range of Fara-ATP concentrations tested (10-50 microM). In contrast, primer RNA formation was stimulated by concentrations of ara-CTP (25-200 microM) and aphidicolin (0.5-5 micrograms/ml) that inhibited DNA synthesis. The primer RNA recovered from cell lysates incubated with either Fara-ATP, ara-CTP, or aphidicolin was of normal length, predominately 11 nucleotides. Fara-ATP was a more potent inhibitor of the polydeoxythymidylate primase activity than of the DNA polymerase alpha/delta activities present in the 100,000 x g supernatants of CCRF-CEM cells. Fara-ATP was a noncompetitive inhibitor of DNA primase with respect to ATP [50% inhibitory concentration, 2.3 +/- 0.3 (SD) microM, Ki = 6.1 +/- 0.3 (SE) microM] and the Km(ATP)/Ki (Fara-ATP) was 25. The 50% inhibitory concentration values of Fara-ATP for DNA polymerases alpha/delta activities on calf thymus DNA were 43 +/- 1.6 (SD) microM and greater than 100 microM with respect to dATP and dTTP. The effects of ara-CTP and aphidicolin on these enzymes were opposite those seen with Fara-ATP, since 50% inhibitory concentrations of either ara-CTP or aphidicolin for DNA polymerases alpha/delta did not inhibit polydeoxythymidylate primase activity. The results provide evidence that fludarabine phosphate blocks DNA synthesis in CCRF-CEM cells through inhibition of primer RNA formation. In contrast, the accumulation of primer RNA and RNA-primed Okazaki fragments that is induced by ara-CTP and aphidicolin could lead to the rereplication and amplification of chromosomal DNA segments.


Assuntos
DNA de Neoplasias/biossíntese , DNA Polimerase Dirigida por DNA/biossíntese , Leucemia/metabolismo , RNA Nucleotidiltransferases/biossíntese , RNA/biossíntese , Fosfato de Vidarabina/análogos & derivados , Afidicolina , Arabinofuranosilcitosina Trifosfato/farmacologia , DNA Primase , Nucleotídeos de Desoxiguanina/farmacologia , Diterpenos/farmacologia , Humanos , Células Tumorais Cultivadas , Fosfato de Vidarabina/farmacologia
5.
Cancer Res ; 60(14): 3790-9, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10919652

RESUMO

The c-myc gene is frequently deregulated and overexpressed in human cancers, and strategies designed to inhibit c-myc expression in cancer cells may have considerable therapeutic value. The purpose of the present work was to characterize the antigene and antiproliferative activity of a triple helix-forming oligonucleotide (TFO) targeted to a homopurine-homopyrimidine sequence in the P2 promoter of the c-myc gene. The TFO was synthesized with phosphorothioate (PS) internucleotide linkages to confer resistance to intra- and extracellular nucleases. This property is required of oligonucleotides designed for in vivo testing and therapeutic applications. The PS-TFO was found to form triplex DNA with affinity and specificity comparable with that of the corresponding phosphodiester TFO, as shown by gel mobility shift and footprinting assays. Fluorescence microscopy and polyacrylamide gel analysis showed that the fluorescein-labeled PS-TFO accumulated in nuclei of CEM leukemia cells and remained intact for at least 72 h. Incubation of CEM cells with PS-TFO reduced c-myc RNA and protein levels. A single exposure of leukemia cells to the PS-TFO was sufficient to induce dose-dependent growth inhibitory effects. Growth inhibition correlated with accumulation of cells in S phase and with induction of cell death by apoptosis. The PS-TFO was also effective in other leukemia and lymphoma cell lines. Control oligonucleotides had minimal effects in all assays. These data indicate that the c-myc-targeted PS-TFO is an effective antigene and antiproliferative agent, with potential for testing in vivo as a novel approach to cancer therapy.


Assuntos
DNA , Leucemia/tratamento farmacológico , Leucemia/genética , Oligonucleotídeos/farmacologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/imunologia , Antígenos/metabolismo , Apoptose/efeitos dos fármacos , Western Blotting , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Núcleo Celular/metabolismo , Meios de Contraste/farmacologia , DNA Complementar/metabolismo , Dimetil Sulfóxido/farmacologia , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Éxons , Fluoresceína/farmacologia , Células HL-60 , Humanos , Linfoma/tratamento farmacológico , Linfoma/genética , Microscopia de Fluorescência , Oligonucleotídeos/farmacocinética , RNA/metabolismo , Fase S/efeitos dos fármacos , Solventes/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas
6.
Cancer Res ; 47(18): 4884-9, 1987 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-3621181

RESUMO

DNA damage caused by methazolastone [an analogue of 5-(3,3-dimethyl-1-triazeno)imidazole-4-carboxamide which does not require metabolic activation] was investigated in L-1210 leukemia which is sensitive to this drug and in a L-1210 subline [L-1210/N,N-bis(2-chloroethyl)-N-nitrosourea (BCNU)] which is resistant to both chloroethylnitrosoureas and methyltriazenes. Both in vitro and in vivo metazolastone caused formation of DNA alkali-labile sites (assessed by alkaline elution techniques) which were present in similar amounts and repaired at a similar rate in L-1210 and L-1210/BCNU. This suggests that these lesions are not crucial to methyltriazenes activity. DNA alkali-labile sites may be due to the removal of 7-methylguanine by 7-methylguanine-DNA glycosylase which showed the same activity in L-1210 and L-1210/BCNU. Flow cytometry studies revealed that in L-1210 but not in L-1210/BCNU methazolastone induced an arrest of cells in SL-G2-M phases. This blockade was delayed, occurring after at least two cell divisions after drug treatment and therefore appeared temporally unrelated to the presence of DNA alkali-labile sites. There was three times more O6-methylguanine-DNA methyltransferase in L-1210/BCNU than in L-1210 suggesting that methylation of O6-guanine is an important lesion for methyltriazenes activity and resistance to this drug may be linked to its repair.


Assuntos
Antineoplásicos/farmacologia , Carmustina/farmacologia , Dano ao DNA , Reparo do DNA , Dacarbazina/análogos & derivados , Leucemia L1210/tratamento farmacológico , Animais , DNA de Neoplasias/análise , Dacarbazina/farmacologia , Resistência a Medicamentos , Citometria de Fluxo , Metilação , Metiltransferases/análise , Camundongos , O(6)-Metilguanina-DNA Metiltransferase , Temozolomida
7.
Cancer Res ; 55(19): 4352-60, 1995 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-7671247

RESUMO

WEHI-3B/NOVO is a cloned murine leukemia cell line selected for resistance to novobiocin that is cross-resistant to the cytotoxic action of etoposide (VP-16) and to a lesser extent to a variety of other topoisomerase II (topo II)-reactive drugs. We have reported previously (Cancer Res. 52: 2782-2790, 1992) that WEHI-3B/NOVO cells exhibit a pronounced decrease in VP-16 induced DNA-topo II cross-link formation compared to the parental WEHI-3B/S cell line in intact cells, in the absence of a significant difference in the P4 unknotting activity of topo II assayed in nuclear extracts. Because the pattern of cross-resistance was suggestive of a topo II-mediated mechanism, we have ascertained whether a change in topo II can account for the multidrug-resistant phenotype of WEHI-3B/NOVO cells. No differences existed between WEHI-3B/S and WEHI-3B/NOVO cells in topo II mRNA and protein levels, as well as in the amount of topo II associated with the nuclear matrix. Neither sensitive nor resistant cells expressed detectable levels of the MDR1 gene; however, VP-16 accumulation in WEHI-3B/NOVO cells was 3-4-fold less than that present in WEHI-3B/S cells, whereas doxorubicin accumulation was the same in both cell lines. Over the first 60 s, no difference existed in the rate of uptake of VP-16 between parental and resistant cells; however, beyond the first 60 s of incubation, [3H]VP-16 accumulated to a greater extent in parental sensitive cells. Thus, an increased rate of efflux of VP-16 was responsible for the lower steady-state concentration of the drug in resistant cells. The efflux Km for VP-16 in WEHI-3B/NOVO cells was 254.7 microM and the Vmax was 10.4 pmol/s/10(7) cells. In the presence of the inhibitors of energy metabolism, sodium azide and deoxyglucose, the efflux of VP-16 was markedly inhibited; readdition of glucose restored the original efflux rate. Northern blot analyses using the human 10.1 probe for the 3'-terminal region of the multidrug-resistance protein (MRP) cDNA revealed a mRNA species of approximately 6 kb in WEHI-3B/NOVO cells but not in WEHI-3B/S cells. Overexpression was associated with amplification of the cognate gene. To ascertain whether the overexpressed gene in WEHI-3B/NOVO cells was the murine MRP or a different member of the same superfamily of ATP-binding ABC cassette transporters, a 341-bp MRP cDNA probe was generated from a murine genomic library.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Trifosfato de Adenosina/farmacologia , Antineoplásicos Fitogênicos/farmacocinética , Resistência a Múltiplos Medicamentos/genética , Etoposídeo/farmacocinética , Leucemia Experimental/metabolismo , Sequência de Bases , Transporte Biológico , DNA Topoisomerases Tipo II/análise , Immunoblotting , Dados de Sequência Molecular , Novobiocina/farmacologia , Matriz Nuclear/enzimologia , Células Tumorais Cultivadas
8.
Int Rev Cytol ; 162A: 539-76, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-8575887

RESUMO

Many nuclear functions, including the organization of the chromatin within the nucleus, depend upon the presence of a nuclear matrix. Nuclear matrix proteins are involved in the formation of chromatin loops, control of DNA supercoiling, and regulation and coordination of transcriptional and replicational activities within individual loops. Various structural and functional components of the nuclear matrix represent potential targets for anticancer agents. Alkylating agents and ionizing radiation interact preferentially with nuclear matrix proteins and matrix-associated DNA. Other chemotherapeutic agents, such as fludarabine phosphate and topoisomerase II-active drugs, interact specifically with matrix-associated enzymes, such as DNA primase and the DNA topoisomerase II alpha isozyme. The interactions of these agents at the level of the nuclear matrix may compromise multiple nuclear functions and be relevant to their antitumor activities.


Assuntos
Antineoplásicos/farmacologia , Matriz Nuclear/efeitos dos fármacos , Animais , Antimetabólitos Antineoplásicos/farmacologia , Antineoplásicos Alquilantes/farmacologia , Cromatina/ultraestrutura , DNA Topoisomerases Tipo II/efeitos dos fármacos , Humanos , Matriz Nuclear/efeitos da radiação , Matriz Nuclear/ultraestrutura , Radiação Ionizante
9.
Biochem Pharmacol ; 62(1): 101-10, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11377401

RESUMO

Previous studies have demonstrated that cytosine arabinoside (araC) induces an accumulation of Okazaki fragments, while fludarabine (FaraA) inhibits Okazaki fragment synthesis. We extended these observations in the present study to provide insights into various mechanisms by which these anticancer drugs affect DNA replication and induce genomic instability in human CEM leukemia cells. Neither araC nor FaraA induced a detectable amount of re-replicated DNA in S-phase cells, which indicated that drug-induced alterations in Okazaki fragment synthesis were not accompanied by DNA re-replication. Synthesis on both leading and lagging DNA strands within the c-myc locus was measured in cells incubated with equitoxic concentrations of araC or FaraA. In araC-treated cells, nascent DNA from the lagging strand was enriched about 5-fold compared with the leading strand. In contrast, FaraA did not induce any replication imbalance. AraC- and FaraA induced changes in the frequency of N-(phosphonacetyl)-l-aspartate (PALA) resistance and the extent of CAD gene amplification were monitored as markers of drug-induced genomic instability. At concentrations that reduced cloning efficiency by 50% (IC(50)), araC increased the frequency of PALA resistance about 4-fold, while FaraA did not have a significant effect on the frequency of PALA resistance. Pretreatment with araC also increased the extent of CAD gene amplification. We propose that the imbalanced DNA synthesis induced by araC leads to the accumulation of Okazaki fragments on the lagging arms and single-stranded DNA regions on the leading arms of replication forks. The formation of these abnormal replication structures was associated with the generation of genomic instability.


Assuntos
Antineoplásicos/farmacologia , Citarabina/farmacologia , DNA de Neoplasias/efeitos dos fármacos , Vidarabina/análogos & derivados , Vidarabina/farmacologia , Replicação do DNA/efeitos dos fármacos , DNA de Neoplasias/biossíntese , Humanos , Leucemia , Células Tumorais Cultivadas
10.
Biochem Pharmacol ; 38(13): 2069-75, 1989 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-2660797

RESUMO

Temozolomide (8-carbamoyl-3-methylimidazo[5,1d]-1,2,3,5-tetrazin-4-(3H)-one), an experimental antitumor agent which spontaneously decomposes to 5-(3,3-methyl-1-triazeno) imidazole-4-carboxamide, the active metabolite of the antineoplastic drug DTIC, causes erythroid differentiation of K562 leukemia cells. The increase in epsilon and gamma globin gene expression after temozolomide treatment does not appear to be due to drug-induced hypomethylation of the genes. In other genes containing many methylated sequences such as the proto-oncogenes c-myc and C-Ha-ras, temozolomide caused no detectable change in methylation. In contrast, in the same genes 5-aza-2'-deoxycytidine induced hypomethylation. Temozolomide caused DNA alkali-labile sites and an arrest of the cell cycle in G2 phase. Ethazolastone (its 3-ethylimidazo analogue) which does not cause differentiation of K562 produced no significant DNA damage and G2 phase blockade. DNA damage rather than hypomethylation may be responsible for induction of differentiation.


Assuntos
DNA de Neoplasias/efeitos dos fármacos , Dacarbazina/análogos & derivados , Genes ras/efeitos dos fármacos , Proto-Oncogenes/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos , Northern Blotting , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Dano ao DNA , Dacarbazina/metabolismo , Dacarbazina/farmacologia , Citometria de Fluxo , Humanos , Cinética , Leucemia Mielogênica Crônica BCR-ABL Positiva , Metilação , Temozolomida , Células Tumorais Cultivadas/citologia
11.
Biochem Pharmacol ; 48(9): 1785-95, 1994 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-7980648

RESUMO

Studies were done to determine (a) the subcellular distribution of the alpha (170 kDa) and beta (180 kDa) isozymes of topoisomerase II, and (b) the extent to which each isozyme forms complexes with DNA in tumor cells incubated with and without VM-26. Western blotting revealed that topoisomerase II beta was highly unstable during cell fractionation. However, preincubation of human CEM leukemia cells with 5-100 microM VM-26 for 30 min protected the beta isozyme from degradation by progressively increasing the amount of this isoform bound to DNA. The amount of topoisomerase II beta detected in nuclei of CEM cells incubated for 30 min with 25 microM VM-26 was 7-fold greater than in nuclei from untreated control cells. VM-26 also had a protective effect on topoisomerase II beta in HL-60 leukemia and WiDR colon carcinoma cells. In contrast, the intercalating agents mitoxantrone and m-AMSA did not protect topoisomerase II beta from degradation during cell fractionation. The stabilization of topoisomerase II beta by VM-26 allowed subsequent studies of the subcellular distribution of the topoisomerase II isozymes. Both isozymes were detected in the nonmatrix (high salt-soluble) fraction of nuclei from CEM cells, but only topoisomerase II alpha was present in the nuclear matrix. VM-26 stabilized binding of the alpha and beta topoisomerase II isoenzymes to nonmatrix DNA and topoisomerase II alpha to matrix DNA. The differences observed in the subnuclear distribution and DNA binding pattern of the topoisomerase II isozymes support the hypotheses that each isozyme has a distinct cellular function, and that both the alpha and beta isozymes are potential targets for VM-26 in intact cells. In addition, the results demonstrated that pretreatment of various cell lines with VM-26 is a useful way to stabilize topoisomerase II beta during cell fractionation.


Assuntos
DNA Topoisomerases Tipo II/metabolismo , DNA/metabolismo , Isoenzimas/metabolismo , Frações Subcelulares/metabolismo , Teniposídeo/farmacologia , Amsacrina/farmacologia , Antígenos de Neoplasias , Núcleo Celular/enzimologia , Proteínas de Ligação a DNA , Desoxirribonuclease I , Humanos , Isoenzimas/antagonistas & inibidores , Matriz Nuclear/enzimologia , Inibidores da Topoisomerase II , Células Tumorais Cultivadas/efeitos dos fármacos
12.
Leuk Res ; 15(1): 19-24, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1997742

RESUMO

We investigated m-AMSA or doxorubicin (Dx) induced DNA single-strand breaks (DNA-SSB) in myeloid leukemia cells obtained from 8 adult patients suffering from AML. Highly purified AML cells were stimulated to proliferate with the addition of the appropriate growth factor (GCT) and exposed to different concentrations of m-AMSA or Dx for 1 or 4 h, respectively. DNA-SSB were determined by alkaline elution techniques. Either the kinetics or the amounts of DNA-SSB caused by both topoisomerase II inhibitors were variable among different cases. By increasing m-AMSA concentrations there was a concomitant increase in DNA-SSB up to a plateau at the highest concentrations. Dx induced DNA-SSB followed a bell shape curve with a decrease in the number of breaks at the highest concentrations that was evident in most cases. The interindividual variability of Dx-induced DNA-SSB was not correlated with intracellular Dx concentrations as assessed by flow cytometry. No correlation was evident between the amount of DNA breaks induced by m-AMSA and that induced by Dx. These data suggest that AML cells derived from different patients are not necessarily cross-sensitive or cross-resistant to topoisomerase II inhibitors with different chemical structures such as amsacrine or anthracyclines.


Assuntos
Amsacrina/farmacologia , Dano ao DNA , DNA de Cadeia Simples/efeitos dos fármacos , Doxorrubicina/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Adulto , Idoso , Feminino , Humanos , Leucemia Mieloide Aguda/genética , Masculino , Pessoa de Meia-Idade
13.
Leuk Res ; 17(11): 977-82, 1993 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7694001

RESUMO

We investigated the cytotoxic activity and some aspects of the mode of action of 5-aza-2'-deoxycytidine (Aza-dC) in 21 primary cultures of leukemic cells freshly obtained from patients with chronic myeloid leukemia (CML) in blast crisis. The cytotoxic potency of Aza-dC was comparable or even greater than that of 1-beta-D-arabinofuranosylcytosine (Ara-C) in most cases, suggesting that this drug has potential in the therapy of blast crisis of CML. Drug incorporation into DNA was evaluated by exposing leukemic cells simultaneously to 3H-Aza-dC at the concentration of 0.1 micrograms/ml and 14C-thymidine (TdR) used as internal standard. Incorporation of Aza-dC into DNA was detectable in all cases. In 17 samples we evaluated the DNA integrity of leukemic cells exposed to Aza-dC using alkaline elution techniques. The drug caused a detectable amount of DNA alkali labile sites (ALS). DNA-ALS increased in cells exposed to Aza-dC concentrations from 0.1 to 1 microgram/ml but did not further increase at 10 micrograms/ml. A plateau in the levels of DNA-ALS was also seen in human K562 cells exposed to increasing concentrations of Aza-dC from 5 to 10 micrograms/ml, whereas in these cells Aza-dC incorporation into DNA increased with increasing Aza-dC concentrations. Therefore, DNA-ALS caused by Aza-dC are not simply the result of the chemical decomposition of azacytosine molecules incorporated into DNA, but are presumably the result of a saturable DNA repair mechanism (e.g., glycosylases) leading to formation of the apyrimidinic sites.


Assuntos
Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Azacitidina/farmacologia , Azacitidina/uso terapêutico , Crise Blástica , Citarabina/farmacologia , DNA de Neoplasias/metabolismo , Decitabina , Humanos , Fatores de Tempo , Células Tumorais Cultivadas
14.
Cancer Chemother Pharmacol ; 48(3): 188-96, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11592339

RESUMO

PURPOSE: Topotecan is a topoisomerase I inhibitor with demonstrated anticancer activity in preclinical and clinical studies. The purpose of the present study was to evaluate drug-drug interactions in therapeutic regimens that would combine topotecan with microtubule-interfering agents, such as Taxol and vinblastine. METHODS: The cytotoxic activities of various drug combinations and schedules of administration were measured in a colon cancer cell line using the MTT assay. Western blot and flow cytometry were performed to determine the effects of Taxol and vinblastine on topoisomerase I and Bcl-xL protein levels and cell cycle distribution. RESULTS: Brief incubation of colon cancer cells with low concentrations of either Taxol or vinblastine increased the efficacy of a subsequent treatment with topotecan. Preincubation of cells with vinblastine or Taxol reduced by 10- to 40-fold the concentration of topotecan necessary to induce a 50% decrease in cell survival. The effects were maximal when the cells were treated for 5 h with microtubule-interfering agents and then incubated for 19 h in drug-free medium before the addition of topotecan. Under these conditions, both Taxol and vinblastine caused an increase in topoisomerase I protein levels, fraction of S phase cells, and extent of Bcl-xL phosphorylation immediately prior to the addition of topotecan. All these factors may contribute to the increased efficacy of topotecan observed with sequential therapy. CONCLUSION: Combinations of topotecan and microtubule-interfering agents result in synergistic anticancer activity when the drugs are administered sequentially. The promising preclinical data presented here encourage clinical testing of these drug combinations using a sequential schedule of administration.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos/farmacologia , Neoplasias do Colo/tratamento farmacológico , Microtúbulos/efeitos dos fármacos , Paclitaxel/farmacologia , Topotecan/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Vimblastina/farmacologia , Western Blotting , Ciclo Celular/efeitos dos fármacos , Neoplasias do Colo/metabolismo , DNA Topoisomerases Tipo I/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Formazans , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sais de Tetrazólio , Inibidores da Topoisomerase I , Células Tumorais Cultivadas/metabolismo , Proteína bcl-X
15.
Adv Enzyme Regul ; 35: 265-81, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7572348

RESUMO

DNA topoisomerase II is a major protein of the nuclear matrix. The enzyme appears to have a central role in both DNA organization and replication. The importance of nuclear matrix topoisomerase II alpha as a target for certain anticancer agents was evaluated in CEM human leukemia cells. Studies were done to determine the extent to which the alpha (170 kDa) and beta (180 kDa) isozymes of topoisomerase II form covalent enzyme-DNA complexes in whole cells and in the nuclear matrix and nonmatrix fractions of CEM cells that are either sensitive or resistant to topoisomerase II-active anticancer agents. Topoisomerase II alpha was detected in both the high salt-soluble (nonmatrix) and matrix fractions of nuclei from parental CEM cells. Most of the matrix topoisomerase II alpha was tightly bound to DNA in cells incubated with VM-26. In contrast, topoisomerase II beta was detected only in the high salt-soluble (nonmatrix) fraction of the nucleus. The subnuclear distribution of the alpha and beta topoisomerase II isozymes in CEM/VM-1 cells resistant to topoisomerase-active drugs was similar to that in drug-sensitive CEM cells. However, the amount and activity of topoisomerase II alpha in nuclear matrices of CEM/VM-1 cells were decreased 3- to 6-fold relative to that of CEM cells. The differences observed in the subnuclear distribution and DNA binding pattern of the topoisomerase II isozymes support the hypotheses that each isozyme has a distinct cellular function. Furthermore, these results provide evidence that topoisomerase II alpha is the nuclear matrix target for VM-26, and that depletion of the nuclear matrix isozyme contributes to cellular resistance to this anticancer agent.


Assuntos
Antineoplásicos/farmacologia , DNA Topoisomerases Tipo II/metabolismo , DNA de Neoplasias/metabolismo , Leucemia/enzimologia , Matriz Nuclear/enzimologia , Teniposídeo/farmacologia , Trifosfato de Adenosina/farmacologia , Núcleo Celular/enzimologia , Resistencia a Medicamentos Antineoplásicos , Humanos , Isoenzimas/metabolismo , Cloreto de Sódio/farmacologia , Inibidores da Topoisomerase II , Células Tumorais Cultivadas
16.
Anticancer Res ; 8(4): 725-31, 1988.
Artigo em Inglês | MEDLINE | ID: mdl-3178163

RESUMO

The hepatic metabolism and biliary excretion of 4-demethoxydaunorubicin (4DDM) was studied in Crl: CD(SD) BR rats by the liver perfusion technique. In the same strains of rats urinary excretion was investigated in vivo. Daunorubicin (DM) was always included for comparison. The drugs and their metabolites were determined in the perfusion medium, in the bile and liver and in the urine by high-performance liquid chromatography with fluorimetric detection. Compared to its analogue DM, 4DDM markedly differed in the metabolic and excretory profile. The cumulative biliary and urinary excretion of 4DDM and the metabolites was quantitatively lower than that of DM (18% vs 36% of the dose) and was consistent with prolonged persistence of 4DDM in plasma in vivo. The extensive carbonyl reduction of 4DDM and DM observed in previous in vivo pharmacokinetic studies was also evident in this study. 13-hydroxy metabolites, daunorubicinol (DMol) and 4-demethoxydaunorubicinol (4DDMol), either as such or after glycosidic cleavage, i.e. 4DDMol aglycone, were present in appreciable amounts in the perfusion medium, bile, liver and urine. In the hepatobiliary system, however, the 13-hydroxy derivative of DM amounted to a much lower fraction than the DM aglycone (17% vs 50% of the total dose), 80% of the total 4DDM dose was accounted for by 4DDMol aglycone. In urine uncleaved DMol or 4DDMol represented more than 75% of the total amount excreted for both drugs. Conjugation, a major step in the excretion of aglycones, seems to play a minor role in the biliary and urinary excretion of 4DDM and 4DDMol.


Assuntos
Antibióticos Antineoplásicos/farmacocinética , Bile/metabolismo , Daunorrubicina/farmacocinética , Idarubicina/farmacocinética , Fígado/metabolismo , Animais , Biotransformação , Daunorrubicina/metabolismo , Daunorrubicina/urina , Idarubicina/metabolismo , Idarubicina/urina , Cinética , Masculino , Perfusão , Ratos , Ratos Endogâmicos
17.
Anticancer Res ; 11(1): 115-21, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-2018345

RESUMO

Using L1210 and a subline resistance to chloroethylnitrosoureas (L1210/BCNU), we found that the resistance to 1-(2-chloroethyl)-1-nitrosourea (CNU) or to diethyl-1-3-(2-chloroethyl)-3-nitrosoureido ethyl phosphonate (fotemustine) can be reversed by a pretreatment with O6-methyl Guanine (O6-mGua) or temozolomide. In L1210/BCNU but not in L1210 the pretreatment with O6mGua caused an increased peak level of CNU-induced DNA-interstrand crosslinks. We then evaluated whether the resistance to BCNU could be counteracted in vivo by i.p. O6mGua treatment of L1210/BCNU bearing mice. The results were negative due to the fact that O6mGua, which was not toxic when given alone, caused a high toxicity when associated with BCNU.


Assuntos
Antineoplásicos/farmacologia , Carmustina/farmacologia , Dacarbazina/análogos & derivados , Etilnitrosoureia/análogos & derivados , Guanina/análogos & derivados , Leucemia L1210/tratamento farmacológico , Compostos de Nitrosoureia/farmacologia , Compostos Organofosforados/farmacologia , Animais , Carmustina/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Resistência a Medicamentos , Etilnitrosoureia/farmacologia , Etilnitrosoureia/uso terapêutico , Guanina/farmacologia , Guanina/uso terapêutico , Cinética , Camundongos , Camundongos Endogâmicos DBA , Compostos de Nitrosoureia/uso terapêutico , Compostos Organofosforados/uso terapêutico , Temozolomida
18.
Ann Ist Super Sanita ; 25(1): 5-9, 1989.
Artigo em Inglês | MEDLINE | ID: mdl-2751197

RESUMO

Some experiences on the relevance of the DNA repair mechanisms in the sensitivity of mammalian cells to alkylating agents are illustrated. Using mouse tumor cell lines we showed an interesting correlation between O6-alkylguanine repair, reduced cross-link formation, and resistance to antineoplastic drugs like chloroethylnitrosoureas. The same lines displayed cross-resistance to a methylating monomethyltriazene indicating a possible role of O6-methylguanine in the toxicity of alkylating agents. In other studies we characterized some modified tissues (nodules) developed during hepatic chemical carcinogenesis in the rat. Such hyperplastic nodules displayed higher DNA repair ability, in keeping with their resistant phenotype against toxic and carcinogenic compounds. Both approaches, using cells in culture or animals, gave interesting hints suggesting future developments which may be useful in the understanding of cell transformation mechanisms as well as in cancer chemotherapy.


Assuntos
Alquilantes/toxicidade , Reparo do DNA/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Transformação Celular Neoplásica/induzido quimicamente , Camundongos , Ratos
19.
Cancer Gene Ther ; 20(5): 326-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23618947

RESUMO

Therapeutic gene silencing is a promising approach for treatment of cancer. Despite substantial efforts, however, only few such therapeutic methods have been clinically tested. The heterogeneity in gene expression profiles among malignant tissues and the dynamic control of gene expression in individual tumors makes identifying universal and effective targets a challenge. Further development of gene silencing therapy requires new approaches to comprehend and manage gene expression in cancer cells. In this study, we proposed and evaluated experimentally a new approach to design multi-gene silencing therapy. Using a simplified model of gene expression control, we show that genes commonly regulated by the same microRNA represent optimal combinations of targets for small hairpin RNA/small interfering RNA-based gene silencing. The proposed method of target gene selection and co-silencing can be explored as an algorithm for personalized cancer gene therapy.


Assuntos
Expressão Gênica/genética , Inativação Gênica , Terapia Genética/métodos , MicroRNAs/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/terapia , Feminino , Perfilação da Expressão Gênica , Humanos , MicroRNAs/metabolismo , Neoplasias Ovarianas/metabolismo , Interferência de RNA
20.
Oncogene ; 32(18): 2315-24, 2324.e1-4, 2013 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-22733135

RESUMO

By integrating gene profiling and immunohistochemical data with functional experiments in cell lines in this study we show for the first time that doublecortin (DCX) domain containing 2 (DCDC2), a protein belonging to the DCX family and involved in neuronal cell migration, is aberrantly expressed in prostate tumors whereas absent in normal prostate. Furthermore, in patients treated with radical prostatectomy, high levels of DCDC2 RNA were significantly associated with increased biochemical relapse (LogRank Mantel-Cox=0.012). Mechanistically, we found that the ETS transcription factor ESE3/EHF, which is expressed in normal prostate and frequently lost in prostate tumors, maintained DCDC2 repressed by binding to a novel identified ETS binding site in the gene promoter. Consistently, in prostate tumors and in cellular models of gain and loss of ESE3/EHF, the expression of DCDC2 and ESE3/EHF were inversely correlated. In prostate cancer cells, DCDC2 colocalized with microtubules and promoted cell migration and resistance to the microtubule-targeting drug taxol. Collectively, this study establishes DCDC2 as a novel ESE3/EHF oncogenic target in prostate cancer. These findings may be relevant for the clinical management of prostate cancer as DCDC2 may signal tumors more prone to relapse and resistant to taxol treatment.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Proteínas Associadas aos Microtúbulos/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Sítios de Ligação , Movimento Celular/genética , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Paclitaxel/farmacologia , Regiões Promotoras Genéticas , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/cirurgia , Valores de Referência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Moduladores de Tubulina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA