Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Publication year range
1.
Cancer Metastasis Rev ; 35(4): 547-573, 2016 12.
Article in English | MEDLINE | ID: mdl-28025748

ABSTRACT

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.


Subject(s)
Breast Neoplasms/pathology , Disease Models, Animal , Animals , Female , Heterografts , Humans , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Translational Research, Biomedical
2.
Br J Cancer ; 114(2): 177-87, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26695443

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Interferon-gamma/drug effects , Receptors, Estrogen/metabolism , STAT1 Transcription Factor/drug effects , Adaptor Proteins, Signal Transducing , Animals , Antigens/drug effects , Antigens/genetics , Antigens/metabolism , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Capecitabine/pharmacology , Carrier Proteins/drug effects , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 3/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/drug effects , Caspase 7/genetics , Caspase 7/metabolism , Cisplatin/pharmacology , Cytokines/drug effects , Cytokines/genetics , Cytokines/metabolism , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Female , Gene Expression Profiling , Humans , Immunohistochemistry , In Situ Hybridization , Interferon-beta/drug effects , Interferon-beta/genetics , Interferon-beta/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myxovirus Resistance Proteins/drug effects , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Neoplasm Transplantation
3.
Curr Opin Oncol ; 26(6): 556-61, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25188472

ABSTRACT

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.


Subject(s)
Breast Neoplasms/drug therapy , Drug Discovery/methods , Xenograft Model Antitumor Assays/methods , Animals , Female , Humans
4.
Breast Cancer Res ; 14(1): R11, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-22247967

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Transcriptome , Animals , Cluster Analysis , Comparative Genomic Hybridization , DNA Copy Number Variations , Female , Genomic Instability , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Transplantation, Heterologous
5.
Med Sci (Paris) ; 28(1): 89-95, 2012 Jan.
Article in French | MEDLINE | ID: mdl-22289836

ABSTRACT

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.


Subject(s)
Neoplasm Metastasis , Animals , Cell Adhesion , Clone Cells/pathology , Disease Progression , Epithelial-Mesenchymal Transition , Evolution, Molecular , Extracellular Matrix/pathology , Forecasting , Humans , Models, Biological , Mutation , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/physiopathology , Neoplasm Metastasis/prevention & control , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplastic Cells, Circulating , Neoplastic Stem Cells/pathology , Stromal Cells/pathology , Tumor Microenvironment
6.
Proc Natl Acad Sci U S A ; 105(34): 12491-6, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18719102

ABSTRACT

Netrins are secreted molecules with roles in axon guidance and angiogenesis. We identified Netrin-4 as a gene specifically overexpressed in VEGF-stimulated endothelial cells (EC) in vitro as well as in vivo. Knockdown of Netrin-4 expression in EC increased their ability to form tubular structures on Matrigel. To identify which receptor is involved, we showed by quantitative RT-PCR that EC express three of the six Netrin-1 cognate receptors: neogenin, Unc5B, and Unc5C. In contrast to Netrin-1, Netrin-4 bound only to neogenin but not to Unc5B or Unc5C receptors. Neutralization of Netrin-4 binding to neogenin by blocking antibodies abolished the chemotactic effect of Netrin-4. Furthermore, the silencing of either neogenin or Unc5B abolished Netrin-4 inhibitory effect on EC migration, suggesting that both receptors are essential for its function in vitro. Coimmunoprecipitation experiments demonstrated that Netrin-4 increased the association between Unc5B and neogenin on VEGF- or FGF-2-stimulated EC. Finally, we showed that Netrin-4 significantly reduced pathological angiogenesis in Matrigel and laser-induced choroidal neovascularization models. Interestingly, Netrin-4, neogenin, and Unc5B receptor expression was up-regulated in choroidal neovessel EC after laser injury. Moreover, Netrin-4 overexpression delayed tumor angiogenesis in a model of s.c. xenograft. We propose that Netrin-4 acts as an antiangiogenic factor through binding to neogenin and recruitment of Unc5B.


Subject(s)
Endothelial Cells/cytology , Membrane Proteins/metabolism , Neovascularization, Pathologic , Nerve Growth Factors/physiology , Receptors, Cell Surface/metabolism , Animals , Cattle , Cell Line, Tumor , Cells, Cultured , Chemotaxis , Female , Humans , Lasers/adverse effects , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/blood supply , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Netrin Receptors , Netrins , Prostatic Neoplasms/pathology , Protein Binding/physiology , Recombinant Proteins/pharmacology , Transplantation, Heterologous , Up-Regulation/genetics
7.
Anticancer Drugs ; 21(10): 927-31, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20827173

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Estramustine/pharmacology , Gastrointestinal Diseases , Prostatic Neoplasms/drug therapy , Taxoids/pharmacology , Vascular Diseases , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Comorbidity , Docetaxel , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Gastrointestinal Diseases/epidemiology , Humans , Male , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/blood , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/epidemiology , Orchiectomy , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/epidemiology , Survival Rate , Vascular Diseases/epidemiology , Venous Thrombosis/chemically induced , Xenograft Model Antitumor Assays
8.
Anticancer Drugs ; 21(1): 25-32, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19823076

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/drug therapy , Small Cell Lung Carcinoma/drug therapy , Topotecan/therapeutic use , Xenograft Model Antitumor Assays , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Etoposide/administration & dosage , Etoposide/therapeutic use , Female , Gene Expression/drug effects , Humans , Ifosfamide/administration & dosage , Ifosfamide/therapeutic use , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Mice , Mice, Nude , Small Cell Lung Carcinoma/enzymology , Small Cell Lung Carcinoma/metabolism , Topotecan/administration & dosage , Treatment Outcome
9.
Clin Cancer Res ; 15(11): 3697-704, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19435839

ABSTRACT

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.


Subject(s)
Cell Proliferation , ErbB Receptors/metabolism , Glioma/pathology , Neovascularization, Pathologic/metabolism , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gefitinib , Glioma/genetics , Glioma/metabolism , Humans , Mice , Mice, Nude , Mutation , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Organ Culture Techniques , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , RNA Interference , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
10.
Exp Cell Res ; 315(17): 3004-13, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19447103

ABSTRACT

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.


Subject(s)
Pluripotent Stem Cells/pathology , Prostatic Neoplasms/pathology , Stromal Cells/pathology , Animals , DNA Primers , Flow Cytometry , Gene Expression Regulation , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , beta 2-Microglobulin/genetics
11.
Sci Transl Med ; 12(531)2020 02 19.
Article in English | MEDLINE | ID: mdl-32075943

ABSTRACT

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.


Subject(s)
Topoisomerase I Inhibitors , Triple Negative Breast Neoplasms , Humans , Irinotecan/pharmacology , Irinotecan/therapeutic use , Nuclear Proteins/metabolism , Retinoblastoma Binding Proteins , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Ubiquitin-Protein Ligases
12.
Int J Cancer ; 124(5): 1103-11, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19058198

ABSTRACT

Lack of hormone dependency in prostate cancers is an irreversible event that occurs through generation of genomic instability induced by androgen deprivation. Indeed, the cytogenetic profile of hormone-dependent (HD) prostate cancer remains stable as long as it received a hormone supply, whereas the profile of hormone-independent (HID) variants acquired new and various alterations. This is demonstrated here using a HD xenografted model of a human prostate cancer, PAC120, transplanted for 11 years into male nude mice and 4 HID variants obtained by surgical castration. Cytogenetic analysis, done by karyotype, FISH, CGH and array-CGH, shows that PAC120 at early passage presents numerous chromosomal alterations. Very few additional alterations were found between the 5th and 47th passages, indicating the stability of the parental tumor. HID variants largely maintained the core of chromosomal alterations of PAC120 - losses at 6q, 7p, 12q, 15q and 17q sites. However, each HID variant displayed a number of new alterations, almost all being specific to each variant and very few shared by all. None of the HID had androgen receptor mutations. Our study indicates that hormone castration is responsible for genomic instability generating new cytogenetic abnormalities susceptible to alter the properties of cancer cell associated with tumor progression, such as increased cell survival and ability to metastasize.


Subject(s)
Genomic Instability , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/genetics , Animals , Chromosome Aberrations , Chromosome Banding , Comparative Genomic Hybridization , Humans , Male , Mice , Receptors, Androgen/genetics
13.
Int J Cancer ; 124(4): 991-7, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19048622

ABSTRACT

The major long-term prognostic factor for breast cancer patients treated by first-line chemotherapy is response to treatment. We have previously shown that complete responses to high doses epirubicin-cyclophosphamide were observed only in human tumors bearing a TP53 mutation. Three xenografted human breast tumors, 2 of them with a TP53 mutation and one of them without, were studied for their immediate response to this drug association. Cell cycle, cellular senescence and cell death were characterized and quantified on tissue section before and after treatment. The TP53 wild-type tumor showed a strong early induction of senescence-like phenotype with overexpression of SA-beta-gal and p21(CIP1). In contrast both TP53 mutated tumors showed no sign of cell cycle arrest or senescence. Conversely, abnormal mitoses strongly increased, only in TP53 mutated tumors. Thus, in these in vivo models, epirubicin-cyclophosphamide treatment induces senescence-like features in TP53 wild-type tumor, likely accounting for cell cycle arrest and subsequent resistance to treatment. Conversely in TP53 mutated tumors, chemotherapy induces mitotic catastrophe and tumor death, accounting for complete response to this association exclusively in patients with TP53 mutated tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Genes, p53 , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mutation , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Cycle , Cellular Senescence , Cyclophosphamide/administration & dosage , DNA Mutational Analysis , DNA, Neoplasm , Epirubicin/administration & dosage , Humans , Mice , Neoplasm Transplantation , Tumor Suppressor Protein p53/genetics
14.
Anticancer Drugs ; 20(10): 932-40, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19741504

ABSTRACT

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.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/therapeutic use , Lung Neoplasms/drug therapy , Taxoids/therapeutic use , Aged , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cisplatin/pharmacology , Docetaxel , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, p53/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , Taxoids/pharmacology , Xenograft Model Antitumor Assays/methods
15.
Cancer Res ; 67(1): 398-407, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17210723

ABSTRACT

Obtaining representative human colon cancer cell lines from fresh tumors is technically difficult. Using 32 tumor fragments from patients with colon cancer, the present study shows that prior xenograft leads to more efficient cell line establishment compared with direct establishment from fresh tumors (P < 0.05). From 26 tumor specimens, we successfully established 20 tumor xenografts in nude mice (77%); among 19 of these xenografts, 9 (47%) led to cell lines, including four from liver metastases. Only 3 of 31 tumor specimens (9.7%) grew immediately in vitro, and all were derived from primary tumors. To compare major phenotypic and genotypic characteristics of human colon cancer cell lines derived from the same tumor fragment using two protocols, the two pairs of cell lines obtained from 2 of 32 tumor fragments were extensively studied. They displayed similar morphology and were able to form compact spheroids. Chemosensitivity to 5-fluorouracil, CPT11, and L-OHP differed between cell lines obtained from patient tumors and those derived from xenografts. Matched cell lines shared a common core of karyotype alterations and distinctive additional chromosomal aberrations. Expression levels of genes selected for their role in oncogenesis evaluated by real-time quantitative PCR were found to be statistically correlated whatever the in vitro culture model used. In conclusion, xenotransplantation in mice of tumor fragments before establishment of cell lines enables generation of more novel human cancer cell lines for investigation of colon cancer cell biology, opening up the opportunity of reproducing the diversity of this disease.


Subject(s)
Cell Line, Tumor , Colonic Neoplasms/pathology , Animals , Cell Growth Processes/physiology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Drug Screening Assays, Antitumor , Gene Expression Profiling , Humans , Karyotyping , Male , Mice , Mice, Nude , Neoplasm Transplantation , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
16.
Mol Cancer Ther ; 7(8): 2498-508, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18687997

ABSTRACT

The targeting of solid tumors requires delivery tools that resist intracellular and extracellular inactivation, and that are taken up specifically by tumor cells. We have shown previously that the recombinant nontoxic B-subunit of Shiga toxin (STxB) can serve as a delivery tool to target digestive tumors in animal models. The aim of this study was to expand these experiments to human colorectal cancer. Tissue samples of normal colon, benign adenomas, colorectal carcinomas, and liver metastases from 111 patients were obtained for the quantification of the expression of the cellular STxB receptor, the glycosphingolipid globotriaosyl ceramide (Gb(3) or CD77). We found that compared with normal tissue, the expression of Gb(3) was strongly increased in colorectal adenocarcinomas and their metastases, but not in benign adenomas. Short-term primary cultures were prepared from samples of 43 patients, and STxB uptake was studied by immunofluorescence microscopy. Of a given tumor sample, on average, 80% of the cells could visibly bind STxB, and upon incubation at 37 degrees C, STxB was transported to the Golgi apparatus, following the retrograde route. This STxB-specific intracellular targeting allows the molecule to avoid recycling and degradation, and STxB could consequently be detected on tumor cells even 5 days after initial uptake. In conclusion, the targeting properties of STxB could be diverted for the delivery of contrast agents to human colorectal tumors and their metastases, whose early detection and specific targeting remains one of the principal challenges in oncology.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/biosynthesis , Colorectal Neoplasms/therapy , Intestines/microbiology , Shiga Toxins/metabolism , Adult , Aged , Aged, 80 and over , Chromatography, Thin Layer , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Male , Middle Aged , Trihexosylceramides/biosynthesis
17.
Oncotarget ; 10(41): 4249-4251, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31289622

ABSTRACT

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

18.
Clin Cancer Res ; 13(13): 3989-98, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17606733

ABSTRACT

PURPOSE: To establish a panel of human breast cancer (HBC) xenografts in immunodeficient mice suitable for pharmacologic preclinical assays. EXPERIMENTAL DESIGN: 200 samples of HBCs were grafted into Swiss nude mice. Twenty-five transplantable xenografts were established (12.5%). Their characterization included histology, p53 status, genetic analysis by array comparative genomic hybridization, gene expression by Western blotting, and quantitative reverse transcription-PCR. Biological profiles of nine xenografts were compared with those of the corresponding patient's tumor. Chemosensitivities of 17 xenografts to a combination of Adriamycin and cyclophosphamide (AC), docetaxel, trastuzumab, and Degarelix were evaluated. RESULTS: Almost all patient tumors established as xenografts displayed an aggressive phenotype, i.e., high-grade, triple-negative status. The histology of the xenografts recapitulated the features of the original tumors. Mutation of p53 and inactivation of Rb and PTEN proteins were found in 83%, 30%, and 42% of HBC xenografts, respectively. Two HBCx had an ERBB2 (HER2) amplification. Large variations were observed in the expression of HER family receptors and in genomic profiles. Genomic alterations were close to those of original samples in paired tumors. Three xenografts formed lung metastases. A total of 15 of the 17 HBCx (88%) responded to AC, and 8 (47%) responded to docetaxel. One ERBB2-amplified xenograft responded to trastuzumab, whereas the other did not. The drug response of HBC xenografts was concordant with that of the patient's tumor in five of seven analyzable cases. CONCLUSIONS: This panel of breast cancer xenografts includes 15 triple-negative, one ER positive and 2 ERBB2 positive. This panel represents a useful preclinical tool for testing new agents and protocols and for further exploration of the biological basis of drug responses.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Drug Screening Assays, Antitumor/methods , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cyclophosphamide/administration & dosage , Disease Models, Animal , Docetaxel , Doxorubicin/administration & dosage , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Nucleic Acid Hybridization , Oligopeptides/administration & dosage , Taxoids/administration & dosage , Trastuzumab
19.
BMC Pharmacol ; 7: 13, 2007 Oct 27.
Article in English | MEDLINE | ID: mdl-17963518

ABSTRACT

BACKGROUND: Using a human small cell lung cancer (SCLC) xenografted in nude mice, we have previously reported enhanced tumor growth inhibition following chemotherapy in combination with imatinib (STI571). We therefore investigated the in vivo impact of imatinib on the pharmacokinetics and efficacy of chemotherapy. METHODS: Two different human tumors were used: SCLC6 small cell lung cancer xenografted in nude mice, and LY-3 EBV-associated human B-cell lymphoma xenografted in SCID mice. Plasma, urine, and fecal concentrations of etoposide (VP16) were determined by a validated high performance liquid chromatography method. Plasma concentrations of ifosfamidewere determined by a validated gas chromatography assay with nitrogen-phosphorus detection. RESULTS: Slight tumor growth inhibition was induced by imatinib administered alone in one in vivo EBV-associated B-cell lymphomatous xenograft. In contrast, an increase of the chemotherapy-induced antitumor effect was observed in the lymphoma model but not in a small cell lung cancer model when mice bearing human xenografted tumors were treated concomitantly by imatinib and chemotherapy. This antitumor effect was not influenced by concomitant administration of fluconazole. The AUC0-3 h (Area Under the concentration-time Curve) of etoposide was increased when mice were treated with etoposide + imatinib due to decreased fecal excretion. In contrast, imatinib did not appear to influence the urinary excretion of etoposide, and concomitant administration of the CYP3A4 inhibitor, fluconazole, with imatinib did not modify the pharmacokinetics of etoposide plus imatinib alone. CONCLUSION: Altogether, these results therefore justify further prospective phase I and II clinical trials with combinations of etoposide-based chemotherapy and imatinib in patients with certain cancers, such as malignant lymphoma, with careful toxicologic monitoring.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Etoposide/pharmacokinetics , Ifosfamide/pharmacokinetics , Lung Neoplasms/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , Benzamides , Carcinoma, Non-Small-Cell Lung/drug therapy , Chromatography, High Pressure Liquid , Drug Synergism , Etoposide/metabolism , Etoposide/therapeutic use , Female , Humans , Ifosfamide/metabolism , Ifosfamide/therapeutic use , Imatinib Mesylate , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Transplantation, Heterologous
20.
Med Sci (Paris) ; 23(12): 1115-23, 2007 Dec.
Article in French | MEDLINE | ID: mdl-18154714

ABSTRACT

Metastatic dissemination represents a leading cause of death in cancer patients. Elucidating the mechanisms of the metastatic process is therefore essential to control it. Since 1988, when the NME (NM23) gene was discovered, several genes specifically suppressing the metastatic potential of tumor cells, have been identified. These metastasis suppressor genes, which exhibit a reduced expression in metastatic tumor cells, are defined by their capacity to suppress metastatic dissemination in vivo without inhibiting primary tumor growth when transfected into metastatic cell lines and injected into experimental animals. Their decreased expression in a subset of human tumor cohorts is associated with a high metastatic potential, thus confirming the data obtained in experimental models. Most of these genes affect key signal transduction pathways, including mitogen-activated protein kinases, Rho-GTPases and G-protein-coupled receptors. These signaling categories control cell-cell and cell-matrix interactions, which are important in monitoring adhesion, invasion and migration properties of metastatic tumor cells. Reduced expression of metastasis suppressor genes is most often due to epigenetic mechanisms, suggesting that their re-expression could constitute a new anti-metastatic therapy. In this paper, we review the literature on metastasis suppressor genes, with a particular focus on NM23.


Subject(s)
Genes, Tumor Suppressor , NM23 Nucleoside Diphosphate Kinases/genetics , Neoplasm Metastasis/genetics , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Humans , Neoplasm Invasiveness/genetics
SELECTION OF CITATIONS
SEARCH DETAIL