Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Clin Genitourin Cancer ; 22(2): 445-453.e1, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38246830

RESUMO

BACKGROUND: There is new interest in platinum-based treatment of patients with metastatic castration resistant prostate cancer (mCRPC), to which a subgroup responds. Although platinum sensitivity is suggested to be associated with aggressive disease features and distinct molecular profiles, identification of responders is a clinical challenge. In this study, we selected patients who displayed PSA progression during cabazitaxel monotherapy, for combined cabazitaxel and carboplatin treatment. METHODS: In this retrospective study, mCRPC patients received carboplatin and cabazitaxel after biochemical progression following at least 2 cabazitaxel monotherapy cycles. We assessed PSA response, Time to PSA Progression (TTpsa) and Time to Radiographic Progression (TTrad). For a subset of patients, mutational analysis of BRCA-1, BRCA-2, ATM, PTEN, P53 and RB1 was performed. RESULTS: Forty-five patients were included, after a median of 4 (3-6) cycles of cabazitaxel monotherapy. Patients received a median of 3 (2-5) cycles of combined cabazitaxel and carboplatin, on which 12 (26.6%) patients had a PSA decline ≥ 50% from baseline. TTpsa was 2 (1-5) months and TTrad 3 (2-6) months. Adverse events were predominantly grade 1-2. Of the 29 (64.4%) patients evaluable for molecular signature, 6 (13.3%) had BRCA1, BRCA2 or ATM mutations and 12 (26.7%) had a PTEN, P53 or RB1 mutations. The occurrence of these mutations was not associated with any clinical outcome measure. CONCLUSIONS: In this study we showed that patients with PSA progression during cabazitaxel monotherapy could benefit from the addition of carboplatin to cabazitaxel, while prospective identification of these patients remains a clinical challenge.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Taxoides , Masculino , Humanos , Carboplatina/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética , Antígeno Prostático Específico , Resultado do Tratamento , Estudos Retrospectivos , Estudos Prospectivos , Proteína Supressora de Tumor p53/genética , Intervalo Livre de Doença , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico
2.
Int J Radiat Oncol Biol Phys ; 114(5): 849-855, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36302495

RESUMO

PURPOSE: The recently developed European Society for Radiotherapy and Oncology (ESTRO)/European Organization for Research and Treatment of Cancer (EORTC) oligometastatic disease (OMD) classification has not been validated in terms of its prognostic significance. This study stratified patients from the phase II SABR-5 trial based on ESTRO/EORTC criteria and compared progression-free survival (PFS) and overall survival (OS) to determine the prognostic significance of the classification scheme. METHODS AND MATERIALS: The SABR-5 trial was a single arm phase II study conducted at the 6 regional cancer centers across British Columbia (BC), Canada, where SABR for oligometastases was only offered on trial. Patients with up to 5 oligometastases (total or not controlled by prior treatment and including induced OMD) underwent SABR to all lesions. Patients were 18 years of age or older, Eastern Cooperative Oncology Group 0 to 2, and life expectancy ≥6 months. PFS and OS were calculated using the Kaplan-Meier method and differences between OMD groups were assessed with log-rank tests. Univariable and multivariable analyses were performed using Cox regression modeling. RESULTS: Between November 2016 and July 2020, 381 patients underwent SABR on trial. Median follow-up was 27 months (interquartile range, 18-36). The most frequent OMD group was de novo OMD (69%), followed by repeat (16%) and induced (13%). OMD groups differed significantly in PFS (P < .001) but not OS (P = .069). The OMD classification was an independent predictor of both PFS (P = .005) and OS (P = .002). Of the 5 classification factors, only chronicity (synchronous, hazard ratio, 0.52; P = .027) and oligoprogression (hazard ratio, 2.05; P = .004) were independently prognostic for OS. CONCLUSIONS: In this large prospective cohort, the ESTRO/EORTC classification was an independent predictor of PFS and OS and should be used to identify specific patient groups for clinical trials. In this trial population, the prognostic power is largely attributable to chronicity and oligoprogression. Simplification of the framework may be possible in the future and allow for greater ease of use; however, further data on underrepresented OMD groups and histologies will be required.


Assuntos
Neoplasias , Radiocirurgia , Humanos , Adolescente , Adulto , Prognóstico , Estudos Prospectivos , Intervalo Livre de Progressão , Radiocirurgia/métodos , Colúmbia Britânica
3.
Prostate Cancer Prostatic Dis ; 24(3): 871-879, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33746212

RESUMO

BACKGROUND: In 2004 docetaxel was the first life-prolonging drug (LPD) registered for metastatic castration-resistant prostate cancer (mCRPC) patients. Between 2011 and 2014 new LPDs for mCRPC (cabazitaxel, abiraterone, enzalutamide, and radium-223) were introduced in the Netherlands. The objective of this study is to assess the impact of the introduction of new LPDs on treatment patterns and overall survival (OS) over time. PATIENTS AND METHODS: CRPC patients diagnosed in the years 2010-2016 in the observational, retrospective CAPRI registry (20 hospitals) were included and followed up to 2018. Two subgroups were analyzed: treatment-naïve patients (subgroup 1, n = 3600) and post-docetaxel patients (subgroup 2, n = 1355). RESULTS: In both subgroups, the use of any LPD increased: from 57% (2010-2011) to 69% (2014-2015) in subgroup 1 and from 65% (2011-2012) to 79% (2015-2016) in subgroup 2. Chemotherapy as first mCRPC-treatment (i.e., docetaxel) and first post-docetaxel treatment (i.e., cabazitaxel or docetaxel rechallenge) decreased (46-29% and 20-9% in subgroup 1 and 2, respectively), while the use of androgen-receptor targeting treatments (ART) increased from 11% to 39% and 46% to 64% in subgroup 1 and 2, respectively. In subgroup 1, median OS (mOS) from diagnosis CRPC increased from 28.5 months to 31.0 months (p = 0.196). In subgroup 2, mOS from progression on docetaxel increased from 7.9 months to 12.5 months (p < 0.001). After multiple imputations of missing values, in multivariable cox-regression analysis with known prognostic parameters, the treatment period was independent significant for OS in subgroup 1 (2014-2015 vs. 2010-2011 with HR 0.749, p < 0.001) and subgroup 2 (2015-2016 vs. 2011-2012 with HR 0.811, p = 0.037). CONCLUSION: Since 2010, a larger proportion of mCRPC patients was treated with LPDs, which was related to an increased mOS.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Quimiorradioterapia/mortalidade , Neoplasias de Próstata Resistentes à Castração/mortalidade , Rádio (Elemento)/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Androstenos/administração & dosagem , Benzamidas/administração & dosagem , Docetaxel/administração & dosagem , Seguimentos , Humanos , Masculino , Nitrilas/administração & dosagem , Feniltioidantoína/administração & dosagem , Prognóstico , Neoplasias de Próstata Resistentes à Castração/patologia , Neoplasias de Próstata Resistentes à Castração/terapia , Estudos Retrospectivos , Taxa de Sobrevida , Taxoides/administração & dosagem
4.
Prostate Cancer Prostatic Dis ; 23(2): 244-251, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31611636

RESUMO

BACKGROUND: Abiraterone acetate is an oral 17α-hydroxylase/C17,20-lyase (CYP17) inhibitor approved for the treatment of metastatic castration-resistant prostate cancer (mCPRC) patients. Previously, a prospective observational trial demonstrated a relationship between abiraterone trough concentrations (Cmin) in plasma and treatment efficacy. The aim of our study was to investigate the exposure-response relationship of abiraterone and its metabolites, and to study if the proposed target for abiraterone of 8.4 ng/mL is feasible in a "real-world" patient cohort. PATIENTS AND METHODS: mCRPC patients who had at least one abiraterone plasma concentration at steady-state were included in this study. Plasma abiraterone and its metabolites levels were analyzed using a validated liquid chromatography-mass spectrometry method. Using calculated Cmin values of abiraterone and its active metabolite Δ(4)-abiraterone (D4A), univariate, and multivariable Cox regression analyses were performed. RESULTS: Sixty-two patients were included in this retrospective analysis, of which 42% were underexposed (mean abiraterone Cmin ≤ 8.4 ng/mL). In multivariable analysis, Cmin ≥ 8.4 ng/mL was associated with longer prostate-specific antigen (PSA) independent progression-free survival (16.9 vs 6.1 months; p = 0.033), which resulted in a hazard ratio of 0.44 (95% confidence interval: 0.23-0.82, p = 0.01). D4A Cmin did not show a relationship with treatment efficacy. CONCLUSION: Our study shows that mCRPC patients with an abiraterone Cmin ≥ 8.4 ng/mL have a better prognosis compared with patients with low Cmin. Monitoring Cmin of abiraterone can help to identify those patients at risk of suboptimal treatment for whom treatment optimization may be appropriate.


Assuntos
Androstenos/uso terapêutico , Biomarcadores Tumorais/sangue , Antígeno Prostático Específico/sangue , Neoplasias de Próstata Resistentes à Castração/sangue , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias de Próstata Resistentes à Castração/patologia , Estudos Retrospectivos , Taxa de Sobrevida , Resultado do Tratamento
5.
Endocr Relat Cancer ; 25(6): R331-R349, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29618577

RESUMO

Androgen receptor (AR) signaling is vital for the normal development of the prostate and is critically involved in prostate cancer (PCa). AR is not only found in epithelial prostate cells but is also expressed in various cells in the PCa-associated stroma, which constitute the tumor microenvironment (TME). In the TME, AR is expressed in fibroblasts, macrophages, lymphocytes and neutrophils. AR expression in the TME was shown to be decreased in higher-grade and metastatic PCa, suggesting that stromal AR plays a protective role against PCa progression. With that, the functionality of AR in stromal cells appears to deviate from the receptor's classical function as described in PCa cells. However, the biological action of AR in these cells and its effect on cancer progression remains to be fully understood. Here, we systematically review the pathological, genomic and biological literature on AR actions in various subsets of prostate stromal cells and aim to better understand the consequences of AR signaling in the TME in relation to PCa development and progression.


Assuntos
Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Microambiente Tumoral , Imunidade Adaptativa , Animais , Fibroblastos Associados a Câncer/metabolismo , Células Endoteliais/metabolismo , Humanos , Imunidade Inata , Masculino
6.
Oncogene ; 37(3): 313-322, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-28925401

RESUMO

Androgen receptor (AR) is a key player in prostate cancer development and progression. Here we applied immunoprecipitation mass spectrometry of endogenous AR in LNCaP cells to identify components of the AR transcriptional complex. In total, 66 known and novel AR interactors were identified in the presence of synthetic androgen, most of which were critical for AR-driven prostate cancer cell proliferation. A subset of AR interactors required for LNCaP proliferation were profiled using chromatin immunoprecipitation assays followed by sequencing, identifying distinct genomic subcomplexes of AR interaction partners. Interestingly, three major subgroups of genomic subcomplexes were identified, where selective gain of function for AR genomic action in tumorigenesis was found, dictated by FOXA1 and HOXB13. In summary, by combining proteomic and genomic approaches we reveal subclasses of AR transcriptional complexes, differentiating normal AR behavior from the oncogenic state. In this process, the expression of AR interactors has key roles by reprogramming the AR cistrome and interactome in a genomic location-specific manner.


Assuntos
Carcinogênese/genética , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Proteínas de Homeodomínio/metabolismo , Neoplasias da Próstata/genética , Receptores Androgênicos/genética , Androgênios/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Epigênese Genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genômica , Fator 3-alfa Nuclear de Hepatócito/genética , Proteínas de Homeodomínio/genética , Humanos , Masculino , Camundongos , Camundongos Nus , Próstata/citologia , Próstata/patologia , Neoplasias da Próstata/patologia , Proteômica , Receptores Androgênicos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Eur J Cancer ; 51(17): 2562-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26278646

RESUMO

INTRODUCTION: The treatment armamentarium for metastatic castration-resistant prostate cancer (mCRPC) has expanded with the introduction of several new therapies. In this treatment continuum, it is unclear whether the efficacy of cabazitaxel is affected by prior novel androgen receptor targeted therapies (ART) such as abiraterone and enzalutamide. In this study, we investigated the influence of prior ART on the efficacy of cabazitaxel in men with mCRPC. PATIENTS AND METHODS: Data from an ongoing multicentre, phase II trial were used comprising 114 men with mCRPC treated with cabazitaxel in the post-docetaxel setting. The primary endpoints of the current analysis were prostate-specific antigen (PSA) response (⩾ 50%), and overall survival (OS). Univariate and multivariable analyses were conducted to investigate the influence of prior ART on the efficacy of cabazitaxel. RESULTS: From the 114 patients included in this analysis, 44 men received prior ART and 70 men did not receive prior ART before treatment with cabazitaxel. PSA response rates while on cabazitaxel treatment were similar in patients with and without prior ART (34% versus 40%, respectively, P = 0.53). Likewise, median OS was not significantly different between men with and without prior ART (13.0 versus 14.0 months, respectively, logrank P = 0.65). In multivariable analysis, the only variables significantly associated with OS were performance status, serum albumin and alkaline phosphatase. CONCLUSION: Our study showed that prior treatment with ART may not influence the efficacy of cabazitaxel in men with mCRPC. With emerging evidence of cross-resistance in the treatment of mCRPC, cabazitaxel provides a good treatment option irrespective of prior ART.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Terapia de Alvo Molecular/métodos , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Receptores Androgênicos/metabolismo , Idoso , Idoso de 80 Anos ou mais , Androstenos/administração & dosagem , Benzamidas , Intervalo Livre de Doença , Docetaxel , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Metástase Neoplásica , Nitrilas , Feniltioidantoína/administração & dosagem , Feniltioidantoína/análogos & derivados , Antígeno Prostático Específico/sangue , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Taxoides/administração & dosagem , Resultado do Tratamento
8.
Eur J Surg Oncol ; 39(4): 365-71, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23375648

RESUMO

AIM: To evaluate the outcome of patients with locally advanced muscle-invasive and/or lymph node positive bladder cancer treated with induction chemotherapy and additional surgery. METHODS: All patients who were treated with induction chemotherapy in our institution between 1990 and 2010, were retrospectively evaluated using an institutional database. Induction chemotherapy consisted of methotrexate, vinblastine, doxorubicin and cisplatin (MVAC), or a combination of gemcitabine with either cisplatin or carboplatin (GC). RESULTS: In total 152 patients were identified, with a mean age of 59 years (range 31-76). One hundred and seven patients (70.4%) received MVAC, 35 patients received GC (23.0%) and 10 patients received GC after initial treatment with MVAC (6.6%). Median follow-up was 68 months (range 4-187 months). Overall 125 patients (82.2%) underwent cystectomy, whereas 12 patients (7.9%) received radiotherapy. Fifteen patients had no local treatment. Median overall survival was 18 months (95%CI 15-23 months). In 37.5% of patients with complete clinical response, residual disease was found at surgery (positive predictive value, PPV 62.5%). Complete pathological response was seen in 26.3% of patients, with a 5 year overall survival (OS) estimate of 54% (39%-74%). For patients with persisting node positive disease after induction chemotherapy and surgery OS was significantly worse (p < 0.0001). CONCLUSIONS: Complete clinical and/or pathological response to induction chemotherapy results in a significant survival benefit. The accuracy of the current clinical response evaluation after induction chemotherapy is limited. Although surgery may be important for staging and prognostic purposes, its role is unclear in node positive disease after induction chemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cistectomia , Quimioterapia de Indução , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/cirurgia , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Intervalo Livre de Doença , Feminino , Humanos , Quimioterapia de Indução/efeitos adversos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Estudos Retrospectivos , Análise de Sobrevida , Resultado do Tratamento , Neoplasias da Bexiga Urinária/patologia
9.
Urol Int ; 84(2): 212-20, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20215828

RESUMO

INTRODUCTION: Pim1 overexpression was found to be associated with more advanced prostate cancer. Interleukin-6 (IL-6) induces Pim1 expression and is often found elevated after androgen-ablative therapy. METHODS: Tumor tissue from 31 selected prostate cancer patients and cell lines were studied immunohistochemically for c-myc, Pim1, STAT3, pSTAT3-Tyr705, and androgen receptor (AR) expression. The effect of Pim1 on androgen dependence in prostate cancer was studied in transgene cell lines. RESULTS: Pim1 expression was increased in high-grade prostate tumors (Gleason >7), irrespective of androgen status, was highest in hormone refractory prostate cancer and correlated to pSTAT3-Tyr705 expression levels. Co-expression of Pim1 and MYC was observed specifically after androgen ablation. Pim1 expression could be induced in LNCaP and PC3 cells by IL-6. Pim1 overexpression in PC3 and PNCaP increased the transcriptional activity of the AR at low levels of dihydrotestosterone. Consequently, Pim1 expression increased LNCaP cell survival specifically at castrate levels of androgen. CONCLUSIONS: Pim1 expression was increased in primary tumor samples after androgen ablation. Pim1 overexpression increased AR transcriptional activity in a ligand-dependent manner in prostate cancer cell lines, resulting in enhanced cell survival at castrate levels of androgen.


Assuntos
Regulação Neoplásica da Expressão Gênica , Interleucina-6/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Androgênios/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Imuno-Histoquímica/métodos , Ligantes , Masculino , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptores Androgênicos/metabolismo , Transgenes
10.
Rev Sci Instrum ; 81(2): 023903, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20192505

RESUMO

III-V nanowires are useful platforms for studying the electronic and mechanical properties of materials at the nanometer scale. However, the costs associated with commercial nanowire growth reactors are prohibitive for most research groups. We developed hot-wall and cold-wall metal organic vapor phase epitaxy reactors for the growth of InAs nanowires, which both use the same gas handling system. The hot-wall reactor is based on an inexpensive quartz tube furnace and yields InAs nanowires for a narrow range of operating conditions. Improvement of crystal quality and an increase in growth run to growth run reproducibility are obtained using a homebuilt UHV cold-wall reactor with a base pressure of 2x10(-9) Torr. A load lock on the UHV reactor prevents the growth chamber from being exposed to atmospheric conditions during sample transfers. Nanowires grown in the cold-wall system have a low defect density, as determined using transmission electron microscopy, and exhibit field effect gating with mobilities approaching 16,000 cm(2)/(V s).

11.
Phys Med Biol ; 55(3): 723-33, 2010 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-20071759

RESUMO

The dosimetric consequences of plans optimized using a commercial treatment planning system (TPS) for hypofractionated radiation therapy are evaluated by re-calculating with Monte Carlo (MC). Planning guidelines were in strict accordance with the Canadian BR25 protocol which is similar to the RTOG 0236 and 0618 protocols in patient eligibility and total dose, but has a different hypofractionation schedule (60 Gy in 15 fractions versus 60 Gy in 3 fractions). A common requirement of the BR25 and RTOG protocols is that the dose must be calculated by the TPS without tissue heterogeneity (TH) corrections. Our results show that optimizing plans using the pencil beam algorithm with no TH corrections does not ensure that the BR25 planning constraint of 99% of the PTV receiving at least 95% of the prescription dose would be achieved as revealed by MC simulations. This is due to poor modelling of backscatter and lateral electronic equilibrium by the TPS. MC simulations showed that as little as 75% of the PTV was actually covered by the 95% isodose line. The under-dosage of the PTV was even more pronounced if plans were optimized with the TH correction applied. In the most extreme case, only 23% of the PTV was covered by the 95% isodose.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/radioterapia , Método de Monte Carlo , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador/métodos , Radioterapia/métodos , Algoritmos , Simulação por Computador , Humanos , Guias de Prática Clínica como Assunto , Estudos Retrospectivos , Espalhamento de Radiação
12.
Ann Oncol ; 20(1): 182-7, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18701427

RESUMO

Glioblastoma multiforme (GBM), the most frequent malignant brain tumor, has a poor prognosis, but is relatively sensitive to radiation. Both gemcitabine and its metabolite difluorodeoxyuridine (dFdU) are potent radiosensitizers. The aim of this phase 0 study was to investigate whether gemcitabine passes the blood-tumor barrier, and is phosphorylated in the tumor by deoxycytidine kinase (dCK) to gemcitabine nucleotides in order to enable radiosensitization, and whether it is deaminated by deoxycytidine deaminase (dCDA) to dFdU. Gemcitabine was administered at 500 or 1000 mg/m(2) just before surgery to 10 GBM patients, who were biopsied after 1-4 h. Plasma gemcitabine and dFdU levels varied between 0.9 and 9.2 microM and 24.9 and 72.6 microM, respectively. Tumor gemcitabine and dFdU levels varied from 60 to 3580 pmol/g tissue and from 29 to 72 nmol/g tissue, respectively. The gene expression of dCK (beta-actin ratio) varied between 0.44 and 2.56. The dCK and dCDA activities varied from 1.06 to 2.32 nmol/h/mg protein and from 1.51 to 5.50 nmol/h/mg protein, respectively. These enzyme levels were sufficient to enable gemcitabine phosphorylation, leading to 130-3083 pmol gemcitabine nucleotides/g tissue. These data demonstrate for the first time that gemcitabine passes the blood-tumor barrier in GBM patients. In tumor samples, both gemcitabine and dFdU concentrations are high enough to enable radiosensitization, which warrants clinical studies using gemcitabine in combination with radiation.


Assuntos
Neoplasias Encefálicas/metabolismo , Desoxicitidina/análogos & derivados , Glioblastoma/metabolismo , Radiossensibilizantes/farmacocinética , Adulto , Idoso , Antimetabólitos Antineoplásicos/sangue , Antimetabólitos Antineoplásicos/farmacocinética , Antimetabólitos Antineoplásicos/uso terapêutico , Disponibilidade Biológica , Biópsia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Citidina Desaminase , Desoxicitidina/sangue , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapêutico , Desoxicitidina Quinase/metabolismo , Feminino , Floxuridina/sangue , Floxuridina/farmacocinética , Floxuridina/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/cirurgia , Humanos , Masculino , Pessoa de Meia-Idade , Nucleosídeo Desaminases/metabolismo , Radiossensibilizantes/uso terapêutico , Gencitabina
13.
Biochem Pharmacol ; 73(10): 1548-57, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17324380

RESUMO

Gemcitabine and ara-C have multiple mechanisms of action: DNA incorporation and for gemcitabine also ribonucleotide reductase (RNR) inhibition. Since dCTP competes with their incorporation into DNA, dCTP depletion can potentiate their cytotoxicity. We investigated whether additional RNR inhibition by Triapine (3-AP), a new potent RNR inhibitor, enhanced cytotoxicity of gemcitabine and ara-C in four non-small-cell-lung-cancer (NSCLC) cell lines, using the multiple-drug-effect analysis. Simultaneous and sequential exposure (preexposure to 3-AP for 24h) in a constant molar ratio of 3-AP and gemcitabine was antagonistic/additive in all cell lines. Preexposure to 3-AP at an IC(25) concentration for 24h before variable concentrations of gemcitabine was synergistic. RNR inhibition by 3-AP resulted in a more synergistic interaction in combination with ara-C, which does not inhibit RNR. Two cell lines with pronounced synergism (SW1573) or antagonism (H460) for gemcitabine/3-AP, were evaluated for accumulation of the active metabolites, dFdCTP and ara-CTP. Simultaneous exposure induced no or a small increase, but ara-CTP increased after pretreatment with 3-AP, 4-fold in SW1573 cells, but not in H460 (<1.5 fold). Ara-C and gemcitabine incorporation into DNA were more pronounced (about 2-fold increased) for sequential treatment in SW1573 compared to H460 cells (<1.5 fold). This was not related to the activity and expression of deoxycytidine kinase and the M2 subunit of RNR. In conclusion, RNR inhibition by 3-AP prior to gemcitabine or ara-C exposure stimulates accumulation of the active metabolites and incorporation into DNA. The combination 3-AP/Ara-C is more synergistic than 3-AP/gemcitabine possibly because gemcitabine already inhibits RNR, but ara-C does not.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Arabinofuranosilcitosina Trifosfato/metabolismo , Citarabina/farmacologia , DNA/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Piridinas/farmacologia , Ribonucleotídeo Redutases/metabolismo , Tiossemicarbazonas/farmacologia , Animais , DNA/metabolismo , Desoxicitidina/metabolismo , Desoxicitidina/farmacologia , Sinergismo Farmacológico , Humanos , Piridinas/metabolismo , Ribonucleotídeo Redutases/efeitos dos fármacos , Tiossemicarbazonas/metabolismo , Células Tumorais Cultivadas , Gencitabina
14.
Nucleosides Nucleotides Nucleic Acids ; 25(9-11): 1001-7, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17065054

RESUMO

To study in detail the relation between gene expression and resistance against gemcitabine, a cell line was isolated from a tumor for which gemcitabine resistance was induced in vivo. Similar to the in vivo tumor, resistance in this cell line, C 26-G, was not related to deficiency of deoxycytidine kinase (dCK). Micro-array analysis showed increased expression of ribonucleotide reductase (RR) subunits M1 and M2 as confirmed by real time PCR analysis (28- and 2.7-fold, respectively). In cell culture, moderate cross-resistance (about 2-fold) was observed to 1-ss-D-arabinofuranosylcytosine (ara-C), 2-chloro-2'deoxyadenosine (CdA), LY231514 (ALIMTA), and cisplatin (CDDP), and pronounced cross-resistance (>23-fold) to 2',2'-difluorodeoxyuridine (dFdU) and 2',2'-difluorodeoxyguanosine (dFdG). Culture in the absence of gemcitabine reduced resistance as well as RRM1 RNA expression, demonstrating a direct relationship of RRM1 RNA expression with acquired resistance to gemcitabine.


Assuntos
Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos , Ribonucleotídeo Redutases/biossíntese , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cladribina/farmacologia , Citarabina/farmacologia , Desoxicitidina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Glutamatos/farmacologia , Guanina/análogos & derivados , Guanina/farmacologia , Humanos , Concentração Inibidora 50 , Pemetrexede , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleotídeo Redutases/química , Gencitabina
15.
Nucleosides Nucleotides Nucleic Acids ; 23(8-9): 1329-33, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15571253

RESUMO

UNLABELLED: Gemcitabine is a deoxycytidine analog, which can be inactivated by deamination catalyzed by deoxycytidine deaminase (dCDA). Altered transport over the cell membrane is a mechanism of resistance to gemcitabine. To facilitate accumulation, the fatty acid derivative CP-4125 was synthesized. Since, the fatty acid is acylated at the site of action of dCDA, a decreased deamination was expected. CP-4125 was equally active as gemcitabine in a panel of rodent and human cell lines and in human melanoma xenografts bearing mice. In contrast to gemcitabine, CP-4125 was not deaminated but inhibited deamination of deoxycytidine and gemcitabine. Pools of the active triphosphate of gemcitabine increased for over 20 hr after CP-4125 exposure, while these pools decreased directly after removal of gemcitabine. IN CONCLUSION: CP-4125 is an interesting new gemcitabine derivative.


Assuntos
Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Ácidos Graxos/metabolismo , Leucemia/tratamento farmacológico , Animais , Antimetabólitos Antineoplásicos/farmacologia , Carbono/química , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células , Citidina Desaminase , DNA/química , Dano ao DNA , Humanos , Concentração Inibidora 50 , Melanoma/patologia , Camundongos , Camundongos Nus , Modelos Químicos , Transplante de Neoplasias , Nucleosídeo Desaminases/metabolismo , Fosforilação , Ratos , Fatores de Tempo , Gencitabina
16.
Nucleosides Nucleotides Nucleic Acids ; 23(8-9): 1523-6, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15571290

RESUMO

Resistance to, the hydrophilic drug ara-C, might be meditated by decreased membrane transport. Lipophilic prodrugs were synthesized to facilitate uptake. These compounds were equally active as ara-C, while the compounds with the shortest fatty-acid group and highest number of double bonds were the more active. These compounds also show a better retention profile, their effect is retained longer than for ara-C.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Citarabina/farmacologia , Desoxicitidina/análogos & derivados , Ácidos Graxos/metabolismo , Leucemia/tratamento farmacológico , Neoplasias/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/farmacologia , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos , Humanos , Concentração Inibidora 50 , Camundongos , Ratos , Fatores de Tempo , Gencitabina
17.
Biochem Pharmacol ; 67(3): 503-11, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15037202

RESUMO

1-beta-D-arabinofuranosylcytosine (ara-C) is a deoxycytidine analog with activity in leukemia, which requires phosphorylation by deoxycytidine kinase (dCK) to allow formation of its active phosphate 1-beta-D-arabinofuranosylcytosine triphosphate, but can be deaminated by deoxycytidine deaminase. Altered membrane transport is also a mechanism of drug resistance. In order to facilitate ara-C uptake and prolong retention in the cell, lipophilic prodrugs were synthesized. Fatty acid groups with a varying acyl chain length and number of double bonds were esterified at the 5' position on the sugar moiety of ara-C. The compounds were tested in two pairs of ara-C resistant leukemic cell lines (murine L1210 and rat BCLO and their resistant variants L4A6 and Bara-C, respectively) and two pairs of cell lines with a resistance to gemcitabine, another deoxycytidine analog (human ovarian cancer A2780 and murine colon cancer C26-A and their resistant variants AG6000 and C26-G, respectively). L4A6, Bara-C and AG6000 have varying degrees of decreased dCK activity, while the mechanism for C26-G is not yet clear. In the parent cell lines, ara-C was more active, but in the resistant variants several of the analogs were more active, while the degree of cross-resistance varied. In AG6000 with a total dCK deficiency, all compounds were inactive. Structure-activity relation analysis showed that ara-C derivatives with shorter acyl chains and more double bonds were more active in the parental and drug resistant cells. Further mechanistic studies were performed with the elaidic acid derivative of ara-C (CP-4055). CP-4055 inhibited deamination of dCyd partly and induced DNA synthesis inhibition effectively in C26-A and C26-G cells, but the retention of inhibition was much longer for CP-4055 than for ara-C. In contrast to ara-C, CP-4055 inhibited RNA synthesis for 60% after drug exposure. In conclusion, CP-4055 seems to be a promising prodrug, whose effects were different and longer lasting than for the parent drug.


Assuntos
Antineoplásicos/farmacologia , Citarabina/farmacologia , Ácidos Graxos/química , Animais , Linhagem Celular Tumoral , Citarabina/análogos & derivados , Citidina Desaminase , DNA/biossíntese , DNA/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Leucemia/patologia , Nucleosídeo Desaminases/metabolismo , RNA/biossíntese , RNA/efeitos dos fármacos , Ratos
18.
Br J Cancer ; 88(12): 1963-70, 2003 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-12799644

RESUMO

Gemcitabine (2',2'-difluorodeoxycytidine) is a deoxycytidine analogue that is activated by deoxycytidine kinase (dCK) to its monophosphate and subsequently to its triphosphate dFdCTP, which is incorporated into both RNA and DNA, leading to DNA damage. Multidrug resistance (MDR) is characterised by an overexpression of the membrane efflux pumps P-glycoprotein (P-gP) or multidrug resistance-associated protein (MRP). Gemcitabine was tested against human melanoma, non-small-cell lung cancer, small-cell lung cancer, epidermoid carcinoma and ovarian cancer cells with an MDR phenotype as a result of selection by drug exposure or by transfection with the mdr1 gene. These cell lines were nine- to 72-fold more sensitive to gemcitabine than their parental cell lines. The doxorubicin-resistant cells 2R120 (MRP1) and 2R160 (P-gP) were nine- and 28-fold more sensitive to gemcitabine than their parental SW1573 cells, respectively (P<0.01), which was completely reverted by 25 micro M verapamil. In 2R120 and 2R160 cells, dCK activities were seven- and four-fold higher than in SW1573, respectively, which was associated with an increased dCK mRNA and dCK protein. Inactivation by deoxycytidine deaminase was 2.9- and 2.2-fold decreased in 2R120 and 2R160, respectively. dFdCTP accumulation was similar in SW1573 and its MDR variants after 24 h exposure to 0.1 micro M gemcitabine, but dFdCTP was retained longer in 2R120 (P<0.001) and 2R160 (P<0.003) cells. 2R120 and 2R160 cells also incorporated four- and six-fold more [(3)H]gemcitabine into DNA (P<0.05), respectively. P-glycoprotein and MRP1 overexpression possibly caused a cellular stress resulting in increased gemcitabine metabolism and sensitivity, while reversal of collateral gemcitabine sensitivity by verapamil also suggests a direct relation between the presence of membrane efflux pumps and gemcitabine sensitivity.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Neoplasias/metabolismo , Divisão Celular/efeitos dos fármacos , Dano ao DNA , Desoxicitidina/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Fosforilação , Células Tumorais Cultivadas , Gencitabina
19.
Int J Oncol ; 22(1): 201-7, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12469205

RESUMO

Gemcitabine (dFdC) and cisplatin (CDDP) act synergistically by an increase in platinum-DNA adduct formation. Since ribonucleotide (NTP) and deoxyribonucleotide (dNTP) levels are essential for DNA-synthesis and repair of DNA damage, we investigated whether disturbances might account for differences in effects between sensitive and resistant cell lines. The human ovarian cancer cell line A2780, its CDDP-resistant variant ADDP and its dFdC-resistant variant AG6000 were exposed for 24 h to dFdC or CDDP alone, or a combination causing moderate to strong growth inhibition. In AG6000 cells UTP levels were 2-fold lower and in ADDP cells almost 2-fold higher than in A2780 cells. Levels of dTTP, dATP and dCTP were 2-5-fold lower in the resistant cell lines. Drug treatment affected NTP and dNTP levels most pronounced in A2780 cells. dFdC alone, at 1.5 nM to 1 micro M increased ATP, GTP and CTP pools 1.2 to 2.0-fold, while 10 micro M dFdC increased UTP 2.5-fold. Combination of dFdC and CDDP increased all NTP levels at low dFdC and CDDP concentrations more than 1.2-fold, but at 20 micro M CDDP only CTP increased 2.4-fold. Only 1.5 nM dFdC increased all dNTP pools more than 1.6-fold, but at 0.1 and 1 micro M dFdC, dATP and dGTP decreased down to 10-fold, while dTTP increased 3-5-fold. CDDP and the combination increased all dNTP pools over 1.4 and 1.9-fold, respectively. In AG6000 cells dFdC and CDDP hardly affected the NTP and dNTP status, except at the high concentrations, which decreased ATP, GTP and UTP levels 1.2-1.8-fold. Both CDDP alone and the combination increased dTTP, dCTP and dATP pools up to 1.6-fold. In ADDP cells NTP and most dNTP levels were hardly affected, except dGTP levels which decreased to non-detectable levels. In conclusion, both dFdC and CDDP induce concentration and combination dependent changes in NTP and dNTP pools.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Cisplatino/administração & dosagem , Desoxicitidina/análogos & derivados , Desoxicitidina/administração & dosagem , Desoxirribonucleotídeos/análise , Neoplasias Ovarianas/tratamento farmacológico , Ribonucleotídeos/análise , Relação Dose-Resposta a Droga , Feminino , Humanos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Células Tumorais Cultivadas , Gencitabina
20.
Biochem Pharmacol ; 61(11): 1401-8, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11331076

RESUMO

Multidrug resistance (MDR), characterized by a cross-resistance to many natural toxin-related compounds, may be caused either by overexpression of a drug efflux pump such as P-glycoprotein, (P-gP), multidrug resistance proteins MRP1-3, or BCRP/MXR or, in the case of DNA topoisomerase II active drugs, by a decrease in the enzymatic activity of the target molecule termed altered topoisomerase MDR (at-MDR). However, human small cell lung carcinoma (SCLC) cell lines showed a collateral sensitivity to 2',2'-difluorodeoxycytidine (gemcitabine, dFdC) and 1-beta-D-arabinofuranosylcytosine (ara-C). H69/DAU, a daunorubicin (DAU)-resistant variant of H69 with a P-gP overexpression, and NYH/VM, a VM-26 (teniposide)-resistant variant of NYH with an at-MDR, were both 2-fold more sensitive to gemcitabine and 7- and 2-fold more sensitive to ara-C, respectively. MDR variants had a 4.3- and 2.0-fold increased activity of deoxycytidine kinase (dCK), respectively. dCK catalyzes the first rate-limiting activation step of both gemcitabine and ara-C. In addition, deoxycytidine deaminase, responsible for inactivation of dFdC and ara-C, was 9.0-fold lower in H69/DAU cells. The level of thymidine kinase 2, a mitochondrial enzyme that can also phosphorylate deoxycytidine and gemcitabine, was not significantly different between the variants. These differences most likely caused an increased accumulation of the active metabolites (dFdCTP, 2.1- and 1.6-fold in NYH/VM and H69/DAU cells, respectively) and of ara-CTP (1.3-fold in NYH/VM cells). Ara-CTP accumulation was not detectable in either H69 variant. The pools of all ribonucleoside and deoxyribonucleoside triphosphates were at least 3- to 4-fold higher in the NYH variants compared to the H69 variants; for dCTP and dGTP this difference was even larger. The higher ribonucleotide pools might explain the >10-fold higher accumulation of dFdCTP in NYH compared to H69 variants. Since dCTP is low, H69 cells might not need a high ara-CTP accumulation to inhibit DNA polymerase. This might be related to the lack of ara-CTP in H69 variants. In addition, the increased CTP, ATP, and UTP pools in the MDR variants might explain the increased ara-CTP and dFdCTP accumulation. In conclusion, the MDR variants of the human SCLC cell lines were collaterally sensitive due to an increased dCK activity, and consequently an increased ara-CTP and dFdCTP accumulation.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Citarabina/farmacologia , Desoxicitidina/farmacologia , Trifosfato de Adenosina/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Arabinofuranosilcitosina Trifosfato/metabolismo , Carcinoma de Células Pequenas , Sobrevivência Celular/efeitos dos fármacos , Citidina Desaminase , Daunorrubicina/farmacologia , Desoxicitidina/análogos & derivados , Desoxicitidina Quinase/metabolismo , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Guanosina Trifosfato/metabolismo , Humanos , Neoplasias Pulmonares , Nucleosídeo Desaminases/metabolismo , Teniposídeo/farmacologia , Timidina Quinase/metabolismo , Células Tumorais Cultivadas , Uridina Trifosfato/metabolismo , Gencitabina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...