Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Sci Transl Med ; 12(531)2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-32075943

RESUMO

Topoisomerase I (TOP1) inhibitors trap TOP1 cleavage complexes resulting in DNA double-strand breaks (DSBs) during replication, which are repaired by homologous recombination (HR). Triple-negative breast cancer (TNBC) could be eligible for TOP1 inhibitors given the considerable proportion of tumors with a defect in HR-mediated repair (BRCAness). The TOP1 inhibitor irinotecan was tested in 40 patient-derived xenografts (PDXs) of TNBC. BRCAness was determined with a single-nucleotide polymorphism (SNP) assay, and expression of Schlafen family member 11 (SLFN11) and retinoblastoma transcriptional corepressor 1 (RB1) was evaluated by real-time polymerase chain reaction (RT-PCR) and immunohistochemistry analyses. In addition, the combination of irinotecan and the ataxia telangiectasia and Rad3-related protein (ATR) inhibitor VE-822 was tested in SLFN11-negative PDXs, and two clinical non-camptothecin TOP1 inhibitors (LMP400 and LMP776) were tested. Thirty-eight percent of the TNBC models responded to irinotecan. BRCAness combined with high SLFN11 expression and RB1 loss identified highly sensitive tumors, consistent with the notion that deficiencies in cell cycle checkpoints and DNA repair result in high sensitivity to TOP1 inhibitors. Treatment by the ATR inhibitor VE-822 increased sensitivity to irinotecan in SLFN11-negative PDXs and abolished irinotecan-induced phosphorylation of checkpoint kinase 1 (CHK1). LMP400 (indotecan) and LMP776 (indimitecan) showed high antitumor activity in BRCA1-mutated or BRCAness-positive PDXs. Last, low SLFN11 expression was associated with poor survival in 250 patients with TNBC treated with anthracycline-based chemotherapy. In conclusion, a substantial proportion of TNBC respond to irinotecan. BRCAness, high SLFN11 expression, and RB1 loss are highly predictive of response to irinotecan and the clinical indenoisoquinoline TOP1 inhibitors.


Assuntos
Inibidores da Topoisomerase I , Neoplasias de Mama Triplo Negativas , Humanos , Irinotecano/farmacologia , Irinotecano/uso terapêutico , Proteínas Nucleares/metabolismo , Proteínas de Ligação a Retinoblastoma , Inibidores da Topoisomerase I/farmacologia , Inibidores da Topoisomerase I/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Ubiquitina-Proteína Ligases
2.
Oncotarget ; 10(41): 4249-4251, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31289622

RESUMO

[This corrects the article DOI: 10.18632/oncotarget.12858.].

3.
Cancer Metastasis Rev ; 35(4): 547-573, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-28025748

RESUMO

Patient-derived xenograft (PDX) models of a growing spectrum of cancers are rapidly supplanting long-established traditional cell lines as preferred models for conducting basic and translational preclinical research. In breast cancer, to complement the now curated collection of approximately 45 long-established human breast cancer cell lines, a newly formed consortium of academic laboratories, currently from Europe, Australia, and North America, herein summarizes data on over 500 stably transplantable PDX models representing all three clinical subtypes of breast cancer (ER+, HER2+, and "Triple-negative" (TNBC)). Many of these models are well-characterized with respect to genomic, transcriptomic, and proteomic features, metastatic behavior, and treatment response to a variety of standard-of-care and experimental therapeutics. These stably transplantable PDX lines are generally available for dissemination to laboratories conducting translational research, and contact information for each collection is provided. This review summarizes current experiences related to PDX generation across participating groups, efforts to develop data standards for annotation and dissemination of patient clinical information that does not compromise patient privacy, efforts to develop complementary data standards for annotation of PDX characteristics and biology, and progress toward "credentialing" of PDX models as surrogates to represent individual patients for use in preclinical and co-clinical translational research. In addition, this review highlights important unresolved questions, as well as current limitations, that have hampered more efficient generation of PDX lines and more rapid adoption of PDX use in translational breast cancer research.


Assuntos
Neoplasias da Mama/patologia , Modelos Animais de Doenças , Animais , Feminino , Xenoenxertos , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Pesquisa Translacional Biomédica
4.
Oncotarget ; 7(47): 77205-77224, 2016 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-27791205

RESUMO

Activation of the IFN/STAT1 pathway is closely associated with drug response and recurrence of breast cancer treated by chemotherapy. The aim of the current study was to elucidate the molecular mechanisms involved upstream and downstream of this pathway in order to identify distinct entities that might be manipulated to improve treatment efficacy. Four breast cancer cell lines (T-47D, MCF7, MDA-MB-231 and HBCx-19 established from the eponymous PDX) were treated in vitro with mafosfamide, a DNA damage inducer. In two of these cell lines (MCF7 and HBCx-19), genotoxic treatment upregulated type I IFN expression leading to paracrine activation of IFN/STAT1 signaling pathway after 6-8 days. We show that STING, a well-characterized inducer of IFN in immune cells, is rapidly triggered in MCF7 cells under genotoxic stress and forms nuclear foci that co-localize with phosphorylated IRF-3 and γH2AX. STING silencing abrogated chemotherapy-induced type I IFN production and signaling and potentiated genotoxic treatment efficacy as it promoted cell death extent and delayed cell colony regrowth. Similar results were obtained after silencing PARP12, one selected gene of the IFN/STAT1 pathway fingerprint. In summary, this study provides the first demonstration of STING activation in breast cancer cells. Our data suggest that genotoxic-induced, STING-mediated type I IFN signaling is a cell-intrinsic mechanism of breast cancer cell survival and regrowth.


Assuntos
Neoplasias da Mama/genética , Ciclofosfamida/análogos & derivados , Proteínas de Membrana/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Ciclofosfamida/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Interferons/metabolismo , Células MCF-7 , Comunicação Parácrina/efeitos dos fármacos , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima
5.
Br J Cancer ; 114(2): 177-87, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26695443

RESUMO

BACKGROUND: Oestrogen receptor-negative (ER-) breast cancer is intrinsically sensitive to chemotherapy. However, tumour response is often incomplete, and relapse occurs with high frequency. The aim of this work was to analyse the molecular characteristics of residual tumours and early response to chemotherapy in patient-derived xenografts (PDXs) of breast cancer. METHODS: Gene and protein expression profiles were analysed in a panel of ER- breast cancer PDXs before and after chemotherapy treatment. Tumour and stromal interferon-gamma expression was measured in xenografts lysates by human and mouse cytokine arrays, respectively. RESULTS: The analysis of residual tumour cells in chemo-responder PDX revealed a strong overexpression of IFN-inducible genes, induced early after AC treatment and associated with increased STAT1 phosphorylation, DNA-damage and apoptosis. No increase in IFN-inducible gene expression was observed in chemo-resistant PDXs upon chemotherapy. Overexpression of IFN-related genes was associated with human IFN-γ secretion by tumour cells. CONCLUSIONS: Treatment-induced activation of the IFN/STAT1 pathway in tumour cells is associated with chemotherapy response in ER- breast cancer. Further validations in prospective clinical trials will aim to evaluate the usefulness of this signature to assist therapeutic strategies in the clinical setting.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Interferon gama/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Fator de Transcrição STAT1/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos/efeitos dos fármacos , Antígenos/genética , Antígenos/metabolismo , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Capecitabina/farmacologia , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspase 3/efeitos dos fármacos , Caspase 3/genética , Caspase 3/metabolismo , Caspase 7/efeitos dos fármacos , Caspase 7/genética , Caspase 7/metabolismo , Cisplatino/farmacologia , Citocinas/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Proteínas do Citoesqueleto/efeitos dos fármacos , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Hibridização In Situ , Interferon beta/efeitos dos fármacos , Interferon beta/genética , Interferon beta/metabolismo , Interferon gama/genética , Interferon gama/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Proteínas Mitocondriais/efeitos dos fármacos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteínas de Resistência a Myxovirus/efeitos dos fármacos , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/metabolismo , Transplante de Neoplasias
6.
Curr Opin Oncol ; 26(6): 556-61, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25188472

RESUMO

PURPOSE OF REVIEW: Patient-derived xenografts (PDXs) are becoming increasing popular as a preclinical tool for evaluating novel therapeutic strategies in cancer. These models maintain the biological characteristics of the donor tumours and have a predictive power in the translation of cancer therapeutics into clinical settings. This review focuses on the rapidly growing body of literature on PDX models of breast cancer and their applications and challenges in cancer drug development. RECENT FINDINGS: Several articles in the last 2 years have reported that breast cancer PDXs can reproduce the phenotype and diversity of patients' tumours. This preservation of breast cancer biology involves a number of different aspects, including gene expression patterns, mutational status, drug response and tumour architecture. These models have been shown to be a valuable tool for the identification of new treatment targets, rational drug combinations, biomarkers and mechanisms of drug resistance. SUMMARY: The development of relevant, predictive models is key to increase the success rate for new drugs. PDX models of breast cancer hold the promise for the development of new and more efficient therapeutic strategies.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Descoberta de Drogas/métodos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Feminino , Humanos
7.
Sci Transl Med ; 6(245): 245ra93, 2014 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-25031268

RESUMO

In vitro studies suggested that sub-millisecond pulses of radiation elicit less genomic instability than continuous, protracted irradiation at the same total dose. To determine the potential of ultrahigh dose-rate irradiation in radiotherapy, we investigated lung fibrogenesis in C57BL/6J mice exposed either to short pulses (≤ 500 ms) of radiation delivered at ultrahigh dose rate (≥ 40 Gy/s, FLASH) or to conventional dose-rate irradiation (≤ 0.03 Gy/s, CONV) in single doses. The growth of human HBCx-12A and HEp-2 tumor xenografts in nude mice and syngeneic TC-1 Luc(+) orthotopic lung tumors in C57BL/6J mice was monitored under similar radiation conditions. CONV (15 Gy) triggered lung fibrosis associated with activation of the TGF-ß (transforming growth factor-ß) cascade, whereas no complications developed after doses of FLASH below 20 Gy for more than 36 weeks after irradiation. FLASH irradiation also spared normal smooth muscle and epithelial cells from acute radiation-induced apoptosis, which could be reinduced by administration of systemic TNF-α (tumor necrosis factor-α) before irradiation. In contrast, FLASH was as efficient as CONV in the repression of tumor growth. Together, these results suggest that FLASH radiotherapy might allow complete eradication of lung tumors and reduce the occurrence and severity of early and late complications affecting normal tissue.


Assuntos
Raios gama , Neoplasias/patologia , Animais , Apoptose/efeitos da radiação , Vasos Sanguíneos/efeitos da radiação , Brônquios/efeitos da radiação , Relação Dose-Resposta à Radiação , Feminino , Humanos , Pulmão/irrigação sanguínea , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
PLoS One ; 8(9): e74179, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24058525

RESUMO

Type 2 diabetes has been associated with decreased risk of prostate cancer in observational studies, and this inverse association has been recently confirmed in several large cohort studies. However the mechanisms involved in this protective effect remain to be elucidated. The aim of the present study was to explore whether different features of type 2 diabetes (hyperinsulinemia, hyperglycemia and tumor necrosis factor alpha [TNF-α]) protect against the development of prostate cancer. For this purpose LNCaP cells were used for in vitro experiments and nude mice in which PAC120 (hormone-dependent human prostate cancer) xenografts had been implanted were used for in vivo examinations. We provide evidence that increasing glucose concentrations downregulate androgen receptor (AR) mRNA and protein levels through NF-κB activation in LNCaP cells. Moreover, there was a synergic effect of glucose and TNFα in downregulating the AR in LNCaP cells. By contrast, insulin had no effect on AR regulation. In vivo experiments showed that streptozotocin-induced diabetes (STZ-DM) produces tumor growth retardation and a significant reduction in AR expression in PAC120 prostate cancer mice. In conclusion, our results suggest that hyperglycemia and TNF-α play an important role in protecting against prostate cancer by reducing androgen receptor levels via NF-κB.


Assuntos
Adenocarcinoma/genética , Diabetes Mellitus Experimental/genética , Glucose/farmacologia , Hiperglicemia/genética , Neoplasias da Próstata/genética , Receptores Androgênicos/genética , Fator de Necrose Tumoral alfa/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma/prevenção & controle , Animais , Linhagem Celular Tumoral , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Regulação da Expressão Gênica , Humanos , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Insulina/farmacologia , Masculino , Camundongos , NF-kappa B/genética , NF-kappa B/metabolismo , Transplante de Neoplasias , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Neoplasias da Próstata/prevenção & controle , Receptores Androgênicos/metabolismo , Transdução de Sinais , Estreptozocina , Transplante Heterólogo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
9.
Mol Cancer Ther ; 11(12): 2693-703, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23012245

RESUMO

Triple-negative breast cancers (TNBC) have an aggressive phenotype with a relatively high rate of recurrence and poor overall survival. To date, there is no approved targeted therapy for TNBCs. Aurora kinases act as regulators of mammalian cell division. They are important for cell-cycle progression and are frequently overexpressed or mutated in human tumors, including breast cancer. In this study, we investigated the therapeutic potential of targeting Aurora kinases in preclinical models of human breast cancers using a pan-inhibitor of Aurora kinases, AS703569. In vitro, AS703569 was tested in 15 human breast cancer cell lines. TNBC cell lines were more sensitive to AS703569 than were other types of breast cancer cells. Inhibition of proliferation was associated with cell-cycle arrest, aneuploidy, and apoptosis. In vivo, AS703569 administered alone significantly inhibited tumor growth in seven of 11 patient-derived breast cancer xenografts. Treatment with AS703569 was associated with a decrease of phospho-histone H3 expression. Finally, AS703569 combined to doxorubicin-cyclophosphamide significantly inhibited in vivo tumor recurrence, suggesting that Aurora kinase inhibitors could be used both in monotherapy and in combination settings. In conclusion, these data indicate that targeting Aurora kinases could represent a new effective approach for TNBC treatment.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Aurora Quinases , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , Imuno-Histoquímica , Células MCF-7 , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
10.
PLoS One ; 7(8): e42252, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22879924

RESUMO

Almost all prostate cancers respond to androgen deprivation treatment but many recur. We postulated that risk of hormone escape--frequency and delay--are influenced by hormone therapy modalities. More, hormone therapies induce crucial biological changes involving androgen receptors; some might be targets for escape prevention. We investigated the relationship between the androgen deprivation treatment and the risk of recurrence using nude mice bearing the high grade, hormone-dependent human prostate cancer xenograft PAC120. Tumor-bearing mice were treated by Luteinizing-Hormone Releasing Hormone (LHRH) antagonist alone, continuous or intermittent regimen, or combined with androgen receptor (AR) antagonists (bicalutamide or flutamide). Tumor growth was monitored. Biological changes were studied as for genomic alterations, AR mutations and protein expression in a large series of recurrent tumors according to hormone therapy modalities. Therapies targeting Her-2 or AKT were tested in combination with castration. All statistical tests were two-sided. Tumor growth was inhibited by continuous administration of the LH-RH antagonist degarelix (castration), but 40% of tumors recurred. Intermittent castration or complete blockade induced by degarelix and antiandrogens combination, inhibited tumor growth but increased the risk of recurrence (RR) as compared to continuous castration (RR(intermittent): 14.5, RR(complete blockade): 6.5 and 1.35). All recurrent tumors displayed new quantitative genetic alterations and AR mutations, whatever the treatment modalities. AR amplification was found after complete blockade. Increased expression of Her-2/neu with frequent ERK/AKT activation was detected in all variants. Combination of castration with a Her-2/neu inhibitor decreased recurrence risk (0.17) and combination with an mTOR inhibitor prevented it. Anti-hormone treatments influence risk of recurrence although tumor growth inhibition was initially similar. Recurrent tumors displayed genetic instability, AR mutations, and alterations of phosphorylation pathways. We postulated that Her-2/AKT pathways allowed salvage of tumor cells under castration and we demonstrated that their inhibition prevented tumor recurrence in our model.


Assuntos
Androgênios/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Tirosina Quinases/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto , Androgênios/deficiência , Animais , Sequência de Bases , Castração , Análise por Conglomerados , Terapia Combinada , Intervalo Livre de Doença , Dosagem de Genes/genética , Humanos , Masculino , Camundongos , Mutação/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Tirosina Quinases/metabolismo , Receptor ErbB-2/genética , Receptores Androgênicos/genética
11.
Med Sci (Paris) ; 28(1): 89-95, 2012 Jan.
Artigo em Francês | MEDLINE | ID: mdl-22289836

RESUMO

The onset of metastasis is a critical event in the natural history of cancer, and is generally associated with a poor clinical outcome. Mechanistically, the metastatic process is made of several steps that are biologically distinct and now rather well characterized. Several explanatory models have been proposed: selective models (clonal selection), adaptive models (initial oncogenesis), involvement of tumor "stem" cells, epithelial-mesenchymal transition… The next progresses are expected to come from the characterization of circulating and disseminated tumor cells, which are two recently opened windows on the metastatic process in patients.


Assuntos
Metástase Neoplásica , Animais , Adesão Celular , Células Clonais/patologia , Progressão da Doença , Transição Epitelial-Mesenquimal , Evolução Molecular , Matriz Extracelular/patologia , Previsões , Humanos , Modelos Biológicos , Mutação , Invasividade Neoplásica/fisiopatologia , Metástase Neoplásica/genética , Metástase Neoplásica/fisiopatologia , Metástase Neoplásica/prevenção & controle , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Células Neoplásicas Circulantes , Células-Tronco Neoplásicas/patologia , Células Estromais/patologia , Microambiente Tumoral
12.
Breast Cancer Res ; 14(1): R11, 2012 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-22247967

RESUMO

INTRODUCTION: Identification of new therapeutic agents for breast cancer (BC) requires preclinical models that reproduce the molecular characteristics of their respective clinical tumors. In this work, we analyzed the genomic and gene expression profiles of human BC xenografts and the corresponding patient tumors. METHODS: Eighteen BC xenografts were obtained by grafting tumor fragments from patients into Swiss nude mice. Molecular characterization of patient tumors and xenografts was performed by DNA copy number analysis and gene expression analysis using Affymetrix Microarrays. RESULTS: Comparison analysis showed that 14/18 pairs of tumors shared more than 56% of copy number alterations (CNA). Unsupervised hierarchical clustering analysis showed that 16/18 pairs segregated together, confirming the similarity between tumor pairs. Analysis of recurrent CNA changes between patient tumors and xenografts showed losses in 176 chromosomal regions and gains in 202 chromosomal regions. Gene expression profile analysis showed that less than 5% of genes had recurrent variations between patient tumors and their respective xenografts; these genes largely corresponded to human stromal compartment genes. Finally, analysis of different passages of the same tumor showed that sequential mouse-to-mouse tumor grafts did not affect genomic rearrangements or gene expression profiles, suggesting genetic stability of these models over time. CONCLUSIONS: This panel of human BC xenografts maintains the overall genomic and gene expression profile of the corresponding patient tumors and remains stable throughout sequential in vivo generations. The observed genomic profile and gene expression differences appear to be due to the loss of human stromal genes. These xenografts, therefore, represent a validated model for preclinical investigation of new therapeutic agents.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Transcriptoma , Animais , Análise por Conglomerados , Hibridização Genômica Comparativa , Variações do Número de Cópias de DNA , Feminino , Instabilidade Genômica , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Transplante Heterólogo
13.
Contemp Clin Trials ; 33(1): 178-83, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21986388

RESUMO

Analysis of preclinical studies using human tumors xenografted into rodents is commonly performed with Tumor Growth Index (TGI) and Tumor Growth Delay index (TGDi). To circumvent the limitations of these parameters, two new parameters, Time To Relapse (TTR) and Tumor Growth Speed (TGS), were developed using a mathematical modeling approach based on an exponential tumor growth. TTR is similar to progression free survival used in human clinical trials and TGS characterizes the pattern of tumor cell proliferation. Parameters were estimated for each rodent by the maximum likelihood method and statistical analyses were performed using ANOVA. These criteria can be used when tumor growths are assessed by repeated measures of their volume. As an example, we used data from a previously published study, which aimed to evaluate the relationship between histology, genetic parameters, and response to alkylating agents in a series of twelve gliomas. The group treated with temozolomide was reanalyzed using our criteria. This group presented a significantly longer TTR than the control group. TTR was also different according to tumor type: oligodendrogliomas relapsed later than glioblastomas. The TGS was different according to the tumor type. Loss of heterozygosity (LOH) 1p ± 19q, LOH 10p ± 10q, telomerase activity, PTEN mutation, and EGFR amplification were related to temozolomide efficacy. Our criteria provide additional information to those given by TGI and TGDi. Due to statistical properties of TTR and TGS, some relations between the parameters such as tumor type or genetic alterations can be studied with TTR and TGS and not with TGI or TGDi.


Assuntos
Biomarcadores Tumorais/genética , Modelos Teóricos , Transplante de Neoplasias , Neoplasias Experimentais , Ensaios Antitumorais Modelo de Xenoenxerto/estatística & dados numéricos , Animais , Terapia Combinada , DNA de Neoplasias/análise , Progressão da Doença , Humanos , Mutação , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Transplante Heterólogo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
14.
Photodiagnosis Photodyn Ther ; 7(4): 275-83, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21112551

RESUMO

PURPOSE: Retinoblastoma is the most common primary intraocular tumor in children. In industrialized countries, 95% of patients are cured by chemotherapy and conservative treatments. However, these treatments can increase the risk of secondary tumors in patients with a constitutional alteration of the RB1 gene. Photodynamic therapy represents a nonmutagenic therapeutic approach, and may reduce the incidence of secondary tumors. To study the in vivo efficacy of photodynamic therapy, human retinoblastoma xenografts were established in nude mice. METHODS: Three xenografted cell lines, RB102-FER, RB109-LAK and RB111-MIL, were characterized and used for therapeutic evaluation. Mice were randomly divided into control and treatment groups with 5-8 mice in each group. Treatment groups received irradiation alone, photosensitizer alone or both in 2 of the 3 models and in the third model, photosensitizer plus irradiation was compared to untreated controls. mTHPC was injected intraperitoneally at a dose of 0.6mg/kg and verteporfin intravenously at a dose of 1mg/kg. Illuminations were performed 24h after mTHPC and 1h after verteporfin injections. RESULTS: A transient but significant response to mTHPC was observed for RB102-FER (p=0.03) and a significant response to mTHPC for RB111-MIL (p<10(-4)) with partial regression maintained for more than 60 days. No significant difference between the different groups was observed for RB109-LAK, except in the verteporfin plus laser group (p=0.01). CONCLUSIONS: The studies confirmed the suitability of the three xenograft models for the evaluation of photodynamic therapy in retinoblastoma. Our findings suggest that PDT may represent an alternative conservative treatment for these tumors.


Assuntos
Mesoporfirinas/uso terapêutico , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Porfirinas/uso terapêutico , Neoplasias da Retina/tratamento farmacológico , Retinoblastoma/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Transplante Heterólogo , Verteporfina
15.
Anticancer Drugs ; 21(10): 927-31, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20827173

RESUMO

Androgen-dependent and castration-resistant prostate cancer (PC) is usually sensitive to docetaxel chemotherapy. Nevertheless, docetaxel resistance frequently appears after several cycles of treatment, raising the problem of salvage treatment for docetaxel-resistant PC patients. Although the combination of docetaxel and estramustine prolongs metastasis-free and overall survival of patients with androgen-independent PC, the use of this modality remains limited in elderly patients or patients with several comorbidities, especially vascular disease or gastrointestinal toxicity, because of unacceptable toxicity including venous thrombosis. The aims of this study were therefore (i) to evaluate the in-vivo efficacy of estramustine combined with docetaxel since initial tumor growth and following the appearance of docetaxel resistance in the androgen-dependent human PC xenograft PAC120, and (ii) to evaluate the efficacy of estramustine in six human androgen-independent PC models derived from PAC120. In docetaxel-resistant tumor-bearing mice, estramustine alone induced a TGD2 of 18 days, whereas the combination of docetaxel and estramustine induced a TGD2 of 50 days (P<0.05) with no significantly different overall survival of mice treated by docetaxel and estramustine since day 1 or since the onset of resistance to docetaxel. Among the six human androgen-independent tumors treated with estramustine alone, two highly sensitive models, two intermediate responding tumors, and two resistant models were observed. Altogether, these results suggest that estramustine should be combined with docetaxel in PC patients, but the use of this treatment could be limited, particularly in elderly patients, to docetaxel-resistant cases.


Assuntos
Antineoplásicos Hormonais/farmacologia , Estramustina/farmacologia , Gastroenteropatias , Neoplasias da Próstata/tratamento farmacológico , Taxoides/farmacologia , Doenças Vasculares , Idoso , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Comorbidade , Docetaxel , Avaliação Pré-Clínica de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Gastroenteropatias/epidemiologia , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias Hormônio-Dependentes/sangue , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/epidemiologia , Orquiectomia , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/sangue , Neoplasias da Próstata/epidemiologia , Taxa de Sobrevida , Doenças Vasculares/epidemiologia , Trombose Venosa/induzido quimicamente , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Anticancer Drugs ; 21(1): 25-32, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19823076

RESUMO

Prediction of human tumor response based on preclinical data could reduce the failure rates of subsequent new anticancer drugs clinical development. Human small-cell lung carcinomas (SCLC) are characterized by high initial sensitivity to chemotherapy but a low median survival time because of drug resistance. The aim of this study was to evaluate the therapeutic relevance of a panel of human SCLC xenografts established in our laboratory using one compromising drug in SCLC, topotecan (TPT). Six SCLC xenografts derived from six patients were used: three were sensitive to a combination of etoposide (VP16), cisplatin (CDDP), and ifosfamide (IFO), and three were resistant, as published earlier. Growth inhibition was greater than 84% for five xenografts at doses of 1-2 mg/kg/day. TPT was combined with IFO, etoposide (VP16), and CDDP. IFO improved the efficacy of TPT in three of the five xenografts and complete responses were obtained even with the less TPT-sensitive xenograft. VP16 increased the efficacy of two of four xenografts and complete responses were obtained. The combination of TPT and CDDP did not improve TPT responses for any of the xenografts tested. Semiquantitative reverse transcriptase-PCR of genes involved in drug response, such as topoisomerase I, topoisomerase IIalpha, multidrug resistance 1 (MDR1), multidrug resistance-associated protein (MRP), lung resistance-related protein (LRP), and glutathione S-transferase pi (GSTpi), did not explain the variability in drug sensitivity between SCLC xenografts. In conclusion, these preclinical data mirror those from published clinical studies suggesting that our panel of SCLC xenografts represents a useful tool for preclinical assessment of new treatments.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Topotecan/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Cisplatino/administração & dosagem , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Etoposídeo/administração & dosagem , Etoposídeo/uso terapêutico , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Ifosfamida/administração & dosagem , Ifosfamida/uso terapêutico , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Carcinoma de Pequenas Células do Pulmão/enzimologia , Carcinoma de Pequenas Células do Pulmão/metabolismo , Topotecan/administração & dosagem , Resultado do Tratamento
17.
PLoS One ; 4(10): e7478, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19829707

RESUMO

We have previously reported that ADP ribosylation factor like 2 (Arl2), a small GTPase, content influences microtubule dynamics and cell cycle distribution in breast tumor cells, as well as the degree and distribution of phosphorylated P53. Here we show, in two different human breast adenocarcinoma models, that Arl2 content has a major impact on breast tumor cell aggressivity both in vitro and in vivo. Cells with reduced content of Arl2 displayed reduced contact inhibition, increased clonogenic or cluster formation as well as a proliferative advantage over control cells in an in vitro competition assay. These cells also caused larger tumors in SCID mice, a phenotype which was mimicked by the in vivo administration of siRNA directed against Arl2. Cells with increased Arl2 content displayed reduced aggressivity, both in vitro and in vivo, with enhanced necrosis and were also found to contain increased PP2A phosphatase activity. A rt-PCR analysis of fresh human tumor breast samples suggested that low Arl2 expression was associated with larger tumor size and greater risk of lymph node involvement at diagnosis. These data underline the role of Arl2, a small GTPase, as an important regulator of breast tumor cell aggressivity, both in vitro and in vivo.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Ligação ao GTP/fisiologia , Regulação Neoplásica da Expressão Gênica , Animais , Linhagem Celular Tumoral , Proliferação de Células , Proteínas de Ligação ao GTP/metabolismo , Humanos , Metástase Linfática , Camundongos , Camundongos SCID , Invasividade Neoplásica , Fenótipo , Fosforilação , Proteína Fosfatase 2/metabolismo , Proteína Supressora de Tumor p53/metabolismo
18.
Anticancer Drugs ; 20(10): 932-40, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19741504

RESUMO

The success of treatment of advanced non-small-cell lung cancer (NSCLC) remains very poor. The aim of this study was, on a series of NSCLC xenografts, to compare the efficacy of standard cisplatin-based or docetaxel-based chemotherapy. Seven human xenografts were obtained from six patients (two xenografts were derived from primary or metastatic tumors of the same patient). Three xenografts were adenocarcinomas and four were squamous cell carcinomas. All xenografts reproduced the same histology as that of the patient's original tumor. Docetaxel, administered as single-agent chemotherapy, induced a significant response in five of the seven NSCLC xenografts (71%), without significant increase after combination with cisplatin, vinorelbine, or gemcitabine. Relative expression of genes putatively involved in drug response was also studied in all xenografts and did not explain the variability of drug sensitivity. In conclusion, this panel of human NSCLC xenografts reliably reproduces the data obtained in patient tumors and the relative sensitivity to docetaxel reported in NSCLC patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Cisplatino/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Taxoides/uso terapêutico , Idoso , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Cisplatino/farmacologia , Docetaxel , Resistencia a Medicamentos Antineoplásicos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes p53/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Taxoides/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
19.
Clin Cancer Res ; 15(11): 3697-704, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19435839

RESUMO

PURPOSE: Epidermal growth factor receptor (EGFR) signal transduction pathways are implicated in malignant glioma aggressiveness and promote tumor cell invasion, proliferation, and angiogenesis. Nevertheless, response to EGFR tyrosine kinase inhibitor gefitinib (Iressa, ZD1839) has been disappointing in clinical trials. One potential explanation may come from the diversity of molecular alterations seen in gliomas. To validate that hypothesis, we have investigated responses to gefitinib on various tumor parameters in human malignant gliomas that exhibited different molecular alterations. EXPERIMENTAL DESIGN: We used a panel of six human malignant gliomas from established xenografts characterized for their genetic (EGFR, PTEN, TP53, and CDKN2A) and molecular (EGFR, PTEN, ERK, and Akt) alterations. Tumors were treated with gefitinib (1 or 10 micromol/L) for prolonged periods (8 or 16 days) in an organotypic brain slice model that allowed quantification of invasion, proliferation, and angiogenesis. RESULTS: In nontreated tumors, EGFR amplification was associated with profuse tumor cell invasion. After treatment, invasion was inhibited in tumors with EGFR amplification in a dose-dependent manner. Treatment had only antiproliferative effect in two of three tumors with EGFR amplification. Tumors with PTEN loss were resistant to treatment. We did not observe shrinkage of the tumors after treatment. None of the tumors had mutations of the EGFR kinase domain. Gefitinib had similar antiangiogenic effect in all of the tumors. CONCLUSIONS: Gefitinib reduces cell invasion in EGFR amplified tumors. PTEN loss of expression seems to be a determinant of resistance. Interestingly, inhibition of angiogenesis by gefitinib seems independent on the EGFR genetic status of the tumors.


Assuntos
Proliferação de Células , Receptores ErbB/metabolismo , Glioma/patologia , Neovascularização Patológica/metabolismo , Animais , Antineoplásicos/farmacologia , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Gefitinibe , Glioma/genética , Glioma/metabolismo , Humanos , Camundongos , Camundongos Nus , Mutação , Invasividade Neoplásica , Neovascularização Patológica/genética , Técnicas de Cultura de Órgãos , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinazolinas/farmacologia , Interferência de RNA , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Exp Cell Res ; 315(17): 3004-13, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19447103

RESUMO

Cancer stem cells are a distinct cellular population that is believed to be responsible for tumor initiation and maintenance. Recent data suggest that solid tumors also contain another type of stem cells, the mesenchymal stem cells or multipotent mesenchymal stromal cells (MSCs), which contribute to the formation of tumor-associated stroma. The Hoechst 33342 efflux assay has proved useful to identify a rare cellular fraction, named Side Population (SP), enriched in cells with stem-like properties. Using this assay, we identified SP cells in a prostate cancer xenograft containing human prostate cancer cells and mouse stromal cells. The SP isolation, subculture and sequential sorting allowed the generation of single-cell-derived clones of murine origin that were recognized as MSC by their morphology, plastic adherence, proliferative potential, adipogenic and osteogenic differentiation ability and immunophenotype (CD45(-), CD81(+) and Sca-1(+)). We also demonstrated that SP clonal cells secrete transforming growth factor beta1 (TGF-beta1) and that their inhibition reduces proliferation and accelerates differentiation. These results reveal the existence of SP cells in the stroma of a cancer xenograft, and provide evidence supporting their MSC nature and the role of TGF-beta1 in maintaining their proliferation and undifferentiated status. Our data also reveal the usefulness of the SP assay to identify and isolate MSC cells from carcinomas.


Assuntos
Células-Tronco Pluripotentes/patologia , Neoplasias da Próstata/patologia , Células Estromais/patologia , Animais , Primers do DNA , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo , Microglobulina beta-2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...